Progress in Management of Advanced Acute Lymphocytic Leukemia in Children

Article Type
Changed
Mon, 08/21/2023 - 12:45
Display Headline
Progress in Management of Advanced Acute Lymphocytic Leukemia in Children

Susan Colace, MD, MSCI
Acute lymphocytic leukemia (ALL) is a heterogeneous malignancy that may develop from B or T lymphocytes (B-ALL, T-ALL) and affects patients of all ages.1 In the United States, an estimated 6,540 new cases are diagnosed each year—including 3,100 in individuals aged < 20 years—and approximately 1,390 deaths annually.2,3 It is the most encountered cancer in patients aged < 20 years, and generally carries a good prognosis; almost all younger patients achieve remission with current therapies. Five-year overall survival (OS) is 90% in patients aged < 15 years, 75% in patients aged 15-19 years, and 61% in adolescent/young adult patients (which generally includes patients up to age 39).2,4,5 In contrast, only about 30% of adults with ALL achieve remission with current therapies.


Incidence peaks in children aged 1-4 years, decreasing thereafter. Cases are highest among Native American/Alaskan Native and Hispanic children, and higher in White than Black children.4 ALL is seen more in patients with certain inherited conditions, including Down syndrome, ataxia telangiectasia, neurofibromatosis type 1, and Bloom syndrome.1

Treatment advances have improved remission rates and outcomes for patients. However, relapse is still a leading cause of death for patients of all ages.6 Prompt diagnosis and care are important to optimize outcomes, as treatment delay is associated with poorer survival.7

Pathophysiology

In ALL, abnormal, immature lymphocytes and progenitor B cells/T cells proliferate uncontrollably and eventually replace healthy cells in bone marrow and the lymphatic system. The loss of healthy cells leads to classic symptoms of cytopenia, splenomegaly, and hepatomegaly.1 B cells and T cells are descended from lymphoid stem cells (and are transformed by germline or somatic mutation into pathogenic cells, leading to symptom development and bone marrow dysfunction. Most pediatric patients have extensive bone marrow involvement at diagnosis, with > 25% blast cells in marrow (defined as M3 disease).4

Presentation

Patients usually present with signs and symptoms that are related to disease-associated anemia, thrombocytopenia, or neutropenia; these signs and symptoms may include fatigue or weakness, pale skin, bleeding or bruising easily, fever or infection, joint or extremity pain, B-cell symptoms such as night sweats or unintentional weight loss, and splenomegaly or hepatomegaly. Central nervous system (CNS) symptoms can include stroke-like symptoms due to leukemic cell invasion of CNS vasculature or neuropathies related to increased intracranial pressure. Sometimes, children may present with no symptoms other than joint or extremity pain.1,3,8

Classification

ALL is classified by whether it derives from B-cell or T-cell progenitor cells and, within these, by typical genetic alterations (Table 1).3,9-15 Some cytogenetics are associated with risk assessment as well. Well-identified B-ALL subtypes include Philadelphia (Ph) chromosome-positive, hyper- and hypodiploidy, and KMT2A rearranged, while newer classifications include Ph-like ALL and B-lymphoblastic leukemia with iAMP21. Provisional T-ALL subtypes include early T-cell precursor lymphoblastic leukemia and natural killer cell lymphoblastic leukemia.3

Table 1. Common Genetic Alterations in ALL

B-cell lineage is present in 88% of pediatric and 75%-80% of adult disease. T-ALL is found in about 12% of pediatric patients and 25% of adults.3,8 Familial syndromes associated with ALL are present in about 4% of pediatric patients, including autosomal dominant germline mutations in RUNX1 (T-cell ALL), ETV6 (B-ALL), PAX5 (B-ALL), IKZF1 (B-ALL and T-ALL), and TP53 (low-hypodiploid ALL).3 If a known-familial genotype is identified, families should be referred for genetic counseling and further testing if needed. If germline mutation is suspected, early identification is important; hereditary ALL can influence treatment choice and use of allogeneic transplantation or radiation.3

A third classification crucial to guiding treatment is Ph-positive vs Ph-negative or Ph-like, the latter strongly associated with abnormal B-cell development due to deletions in related genes.3,16 About 3% to 5% of pediatric patients and 25% of adults have Ph-positive ALL.17 The remission failure rate among pediatric patients treated with chemotherapy was 11% in one study, vs 2%-3% among patients with Ph-negative ALL.10

Diagnosis and Risk Stratification

Diagnosis is based on presentation and molecular features, requiring demonstration of ≥ 20% lymphoblasts in bone marrow biopsy or aspirate or ≥ 1,000 circulating lymphoblasts/mL in peripheral blood. Testing can include immunophenotyping using flow cytometry, molecular characterization of baseline leukemic clone, morphology using hematoxylin and eosin staining and Wright/Giemsa staining, and karyotyping.1,3 CNS involvement is assessed using a lumbar spinal tap.1

Risk stratification is based on molecular features (eg, high- and low-risk mutations, Table 1),3,9-15 which are assessed using fluorescence in-situ hybridization, broad-panel next-generation sequencing, and reverse-transcriptase polymerase chain reaction of bone marrow or peripheral blood.Other risk factors include age, CNS involvement, white blood cell (WBC) count, and response to initial induction or consolidation therapy.3

Pediatric patients are assigned standard or high risk based on factors identified by the Children’s Oncology Group and National Comprehensive Cancer Network (NCCN). Patients
aged 1 to < 10 years with WBC < 50 × 109/L are considered standard risk, and all others are considered high risk. Patients with ALL before age 1 have very high risk. All pediatric patients with T-ALL are considered high risk.3 Ph-positive, Ph-like, hypoploidy, failure to achieve remission with induction, and extramedullary disease are high-risk factors as well, whereas hyperploidy and certain mutations convey low risk.3

Newer treatment strategies for initial ALL diagnosis include targeted therapies. One goal of targeted therapy is avoidance of long-term toxicity, leading to improved survival outcomes. Well-studied targeted therapies include the tyrosine kinase inhibitors used in first-line and subsequent treatment of Ph-positive ALL.3

Treatment Options in Relapsed/Refractory ALL

The initial treatment goal is complete remission (CR) defined as minimal residual disease (MRD) < 0.01% on flow cytometry (Table 2).3 Prognosis is dependent on time and location of relapse. Early relapse (< 18 months from diagnosis) predicts poor survival. Relapse in bone marrow is associated with poorer prognosis than relapse in CNS.11-18 Where possible, consolidation with allogeneic hematopoietic cell transplantation improves survival for patients with early relapse.6 Three approaches have advanced treatment options for relapsed/refractory (R/R) B-ALL, all based around common cell markers seen in B-ALL.

Table 2. Response Criteria in ALL

The CD22-directed antibody-drug conjugate inotuzumab ozogamicin is approved for adults with R/R B-ALL. In clinical trials, a higher percentage of patients had results below the MRD threshold, and longer progression-free survival and OS compared with standard care.19,20

Blinatumomab is a bispecific T-cell engager that binds to CD19 on the surface of B-ALL cells and to CD3 on T cells to trigger apoptosis.21 It was first approved for R/R ALL in adults or children, and is also now approved for treatment in remission with MRD ≥ 0.1%. Patients must demonstrate CD19-positive disease to qualify.15-22 For R/R ALL, blinatumomab improves OS and CR rates compared with standard chemotherapy.23

The use of CAR T-cell therapies has expanded greatly with increasing knowledge about their efficacy and safety. In R/R ALL, tisagenlecleucel (tisa-gen) is approved for treatment of patients aged ≤ 25 years, and brexucabtagene autoleucel (brexucel) is approved for treatment of adults.3,24,25 Patients undergoing the CAR T-cell process have apheresis to collect T cells, which are then manufactured before being reinfused into the patient. Depending on local capabilities, the time between T-cell harvest and reinfusion can extend to weeks.3,26,27 Cytoreduction with CAR T-cell therapy can allow previously ineligible patients (due to bulky disease) to undergo transplant. Patients treated in key clinical trials with tisa-gen or brexu-cel achieved high overall remission rates and improved event-free survival and OS rates compared with historical experience.25,28,29 Important toxicities with CAR T-cell therapy are cytokine release syndrome (CRS) and neurotoxicity, which can develop rapidly. NCCN recommends hospitalizing patients at the first sign of either adverse event. Patients can be managed with tocilizumab or steroids for low-grade CRS or steroids for neurotoxicity. The Society for Immunotherapy of Cancer, American Society of Clinical Oncology, and NCCN have guidelines on management of toxicities related to CAR T-cell therapy as well as management of symptoms and other adverse effects of CRS.5,23,24

Programs also incorporate telemedicine for symptom monitoring and follow-up.32-34 Centers providing CAR T-cell therapy must have a certified Risk Evaluation and Mitigation Strategy (REMS), which ensures adherence to specific guidelines for administration, adverse event management, and patient education.35,36 Overcoming technical, social, and financial barriers to CAR T-cell therapy is an ongoing challenge of great interest.37

R/R T-Cell Precursor ALL

Patients with R/R T-ALL have poor prognosis, partly due to limited treatment options. Nelarabine, a nucleoside analog, is the only approved treatment for R/R T-ALL, but has increasingly been used in first-line therapy added to multiagent chemotherapy as a consolidation and maintenance approach to pediatric disease.3,38,39 Four-year DSF in pediatric patients with newly diagnosed T-ALL undergoing treatment incorporating nelarabine was 88.9%.39 Treatment is associated with grade ≥ 3 neurotoxicity in > 10% of patients, and can include CNS toxicity as well as neuropathy.3

In a recently completed phase 2 trial (NCT03384654), daratumumab was added to standard chemotherapy (vincristine, prednisone, PEG-asparaginase, doxorubicin) for R/R T-ALL in pediatric (ages 1-17 years) and young adult patients (age ≥ 18 years).40 Among 24 pediatric patients, CR was 41.7% and overall response rate (ORR; ORR = CR + CRi) was 83% after 1 cycle of treatment. Ten (41.7%) pediatric patients achieved MRD-negative status as well. ORR was 60% in the 5 older patients. All pediatric patients had at least 1 grade ≥ 3 toxicity, but none of the adverse events led to discontinuation.40

Success in achieving MRD-negative responses in patients treated for R/R ALL has increased interest in using targeted therapies for newly diagnosed patients. Recommended treatment approaches are summarized in Table 3.3

Table 3. Recommended Therapy for R/R ALL

Long-Term Follow-Up and Survivorship

A study of > 500 pediatric patients followed for an average 23 years reassuringly found low prevalence of adverse outcomes related to disease or treatment. Major adverse outcomes such as death due to late relapse; secondary malignancy; or development of osteoporosis, cataracts, and diminished functional status were infrequent.41 Most prevalent were growth effects (short stature or growth hormone insufficiency), likely related to certain treatment approaches.41 Guidelines for long-term follow-up of pediatric patients are available from the Children’s Oncology Group.42

A 2017 systematic review concluded that the quality of life for survivors is diminished upon treatment, and persistently over time for some patients.43 In contrast, a 2022 comparison of long-term survivors (median 20.5 years since diagnosis) of pediatric ALL with healthy controls found that survivors had better quality of life in some domains, including general health, vitality, and mental health.44 Smaller percentages of survivors rated themselves happiest about sleep quality, absence of pain, and physical abilities.44

As therapy patterns and options evolve, continued follow-up is important to ensure patients derive optimal benefit from treatment and post-treatment life.

Click to read more from 2023 Rare Diseases Report: Cancers

References
  1. Puckett Y, Chan O. Acute lymphocytic leukemia. StatPearls [Internet]. Treasure Island, FL: StatPearls Publishing; 2022. Updated June 27, 2022. Accessed April 10, 2023. https://www.ncbi.nlm.nih.gov/books/NBK459149/
  2. Cancer facts & figures 2023. American Cancer Society. 2023. Accessed April 10, 2023. https://www.cancer.org/content/dam/cancer-org/research/cancer-facts-and-statistics/annual-cancer-facts-and-figures/2023/2023-cancer-facts-and-figures.pdf
  3. National Comprehensive Cancer Network. NCCN clinical practice guidelines in oncology: acute lymphoblastic leukemia. Version 1.2022. April 4, 2022. Accessed April 10, 2023. https://www.nccn.org/professionals/physician_gls/pdf/all.pdf
  4. Childhood acute lymphoblastic leukemia (PDQ)—Health Professional Version. National Cancer Institute. Updated February 16, 2023. Accessed April 10, 2023. https://www.cancer.gov/types/leukemia/hp/child-all-treatment-pdq
  5. National Comprehensive Cancer Network. NCCN clinical practice guidelines in oncology: management of immunotherapy-related toxicities. Version 1.2023. March 10, 2023. Accessed April 10, 2023. https://www.nccn.org/professionals/physician_gls/pdf/immunotherapy.pdf
  6. DuVall AS, Sheade J, Anderson D, et al. Updates in the management of relapsed and refractory acute lymphoplastic leukemia: an urgent plea for new treatments is being answered! JCO Oncol Pract. 2022;18(7):479-487. doi:10.1200/OP.21.00843
  7. Baker JM, To T, Beyene J, Zagorski B, Greenberg ML, Sung L. Influence of length of time to diagnosis and treatment on the survival of children with acute lymphoblastic leukemia: a population-based study. Leuk Res. 2014;38(2):204-209. doi:10.1016/j.leukres.2013.11.014
  8. Acute adult lymphoblastic leukemia (PDQ)—Health Professional Version. National Cancer Institute. Updated February 24, 2023. Accessed April 10, 2023. https://www.cancer.gov/types/leukemia/hp/adult-all-treatment-pdq
  9. Trinquand A, Tanguy-Schmidt A, Ben Abdelali R, et al. Toward a NOTCH1/FBXW7/RAS/PTEN–based oncogenetic risk classification of adult T-cell acute lymphoblastic leukemia: a Group for Research in Adult Acute Lymphoblastic Leukemia Study. J Clin Oncol. 2013;31(34):4333-4342. doi:10.1200/JCO.2012.48.5292
  10. Callens C, Baleydier F, Lengline E, et al. Clinical impact of NOTCH1 and/or FBXW7 mutations, FLASH deletion, and TCR status in pediatric T-cell lymphoblastic lymphoma. J Clin Oncol. 2012;30(16):1966-1973. doi:10.1200/JCO.2011.39.7661
  11. Gao C, Liu SG, Zhang RD, et al. NOTCH1 mutations are associated with favourable long-term prognosis in paediatric T-cell acute lymphoblastic leukaemia: a retrospective study of patients treated on BCH-2003 and CCLG-2008 protocol in China. Br J Haematol. 2014;166(2):221-228. doi:10.1111/bjh.12866
  12. Yang YL, Hsiao CC, Chen HY, et al. Absence of biallelic TCRγ deletion predicts induction failure and poorer outcomes in childhood T-cell acute lymphoblastic leukemia. Pediatr Blood Cancer. 2012;58(6):846-851. doi:10.1002/pbc.24021
  13. Gutierrez A, Dahlberg SE, Neuberg DS, et al. Absence of biallelic TCRgamma deletion predicts early treatment failure in pediatric T-cell acute lymphoblastic leukemia. J Clin Oncol. 2010;28(24):3816-3823. doi:10.1200/JCO.2010.28.3390
  14. Bandapalli OR, Zimmermann M, Kox C, et al. NOTCH1 activation clinically antagonizes the unfavorable effect of PTEN inactivation in BFM-treated children with precursor T-cell acute lymphoblastic leukemia. Haematologica. 2013;98(6):928-936. doi:10.3324/haematol.2012.073585
  15. Palmi C, Savino AM, Silvestri D, et al. CRLF2 over-expression is a poor prognostic marker in children with high risk T-cell acute lymphoblastic leukemia. Oncotarget. 2016;7(37):59260-59272. doi:10.18632/oncotarget.10610
  16. Den Boer ML, van Slegtenhorst M, De Menezes RX, et al. A subtype of childhood acute lymphoblastic leukaemia with poor treatment outcome: a genome-wide classification study. Lancet Oncol. 2009;10(2):125-134. doi:10.1016/S1470-2045(08)70339-5
  17. Aricò M, Schrappe M, Hunger SP, et al. Clinical outcome of children with newly diagnosed Philadelphia chromosome-positive acute lymphoblastic leukemia treated between 1995 and 2005. J Clin Oncol. 2010;28(31):4755-4761. doi:10.1200/JCO.2010.30.1325
  18. Nguyen K, Devidas M, Cheng SC, et al.; Children’s Oncology Group. Factors influencing survival after relapse from acute lymphoblastic leukemia: a Children’s Oncology Group study. Leukemia. 2008;22(12):2142-2150. doi:10.1038/leu.2008.251
  19. Besponsa. Prescribing information. Wyeth Pharmaceuticals Inc; 2017. BESPONSA® (inotuzumab ozogamicin) Dosing & Administration |Safety Info (pfizerpro.com)
  20.  Kantarjian HM, DeAngelo DJ, Stelljes M, et al. Inotuzumab ozogamicin versus standard therapy for acute lymphoblastic leukemia. N Engl J Med. 2016;375(8):740-753. doi:10.1056/NEJMoa1509277
  21. Lv M, Liu Y, Liu W, Xing Y, Zhang S. Immunotherapy for pediatric acute lymphoblastic leukemia: recent advances and future perspectives. Front Immunol. 2022;13:921894. doi:10.3389/fimmu.2022.921894
  22. Blincyto. Prescribing information. Amgen; 2022. https://www.pi.amgen.com/-/media/Project/Amgen/Repository/pi-amgen-com/Blincyto/blincyto_pi_hcp_english.pdf
  23. Kantarjian H, Stein A, Gökbuget N, et al. Blinatumomab versus chemotherapy for advanced acute lymphoblastic leukemia. N Engl J Med. 2017;376(9):836-847. doi:10.1056/NEJMoa1609783
  24. Maude SL, Laetsch TW, Buechner J, et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 2018;378(5):439-448. doi:10.1056/NEJMoa1709866
  25. Shah BD, Ghobadi A, Oluwole OO, et al. KTE-X19 for relapsed or refractory adult B-cell acute lymphoblastic leukaemia: phase 2 results of the single-arm, open-label, multicentre ZUMA-3 study. Lancet. 2021;398(10299):491-502. doi:10.1016/S0140-6736(21)01222-8
  26. Bhaskar ST, Dholaria BR, Singsayadeth S, Savani BN, Oluwole OO. Role of bridging therapy during chimeric antigen receptor T cell therapy. EJHaem. 2021;3(suppl 1):39-45. doi:10.1002/jha2.335
  27. Granroth G, Rosenthal A, McCallen M, et al. Supportive care for patients with lymphoma
    undergoing CAR-T-cell therapy: the advanced practice provider’s perspective. Curr Oncol Rep. 2022;24(12):1863-1872. doi:10.1007/s11912-022-01330-z
  28. Laetsch TW, Maude SL, Rives S, et al. Three-year update of tisagenlecleucel in pediatric and young adult patients with relapsed/refractory acute lymphocytic leukemia in the ELIANA trial. J Clin Oncol. 2023;41(9):1664-1669. doi:10.1200/JCO.22.00642
  29. Shah BD, Ghobadi A, Oluwole OO, et al. Two-year follow-up of KTE-X19 in patients with relapsed or refractory adult B-cell acute lymphoblastic leukemia in ZUMA-3 and its contextualization with SCHOLAR-3, an external historical control study. J Hematol Oncol. 2022;15(1):170. doi:10.1186/s13045-022-01379-0
  30. Maus MV, Alexander S, Bishop MR, et al. Society for Immunotherapy of Cancer (SITC) clinical practice guideline on immune effector cell-related adverse events. J Immunother Cancer. 2020;8(2):e001511. doi:10.1136/jitc-2020-001511
  31. Santomasso BD, Nastoupil LJ, Adkins S, et al. Management of immune-related adverse events in patients treated with chimeric antigen receptor T-cell therapy: ASCO Guideline. J Clin Oncol. 2021;39(35):3978-3992. doi:10.1200/JCO.21.01992
  32. Borogovac A, Keruakous A, Bycko M, et al. Safety and feasibility of outpatient chimeric antigen receptor (CAR) T-cell therapy: experience from a tertiary care center. Bone Marrow Transpl. 2022;57(6):1025-1027. doi:10.1038/s41409-022-01664-z
  33. LeBar K, Murawski S, Umayam S, Quinn V. The role of advanced practice providers and telemedicine in reinventing care: the transition of a CAR T-cell transplantation program to the outpatient setting. J Adv Pract Oncol. 2020;11(7):757-763. doi:10.6004/jadpro.2020.11.7.8
  34. Myers GD, Verneris MR, Goy A, Maziarz RT. Perspectives on outpatient administration of CAR-T cell therapy for aggressive B-cell lymphomas and acute lymphoblastic leukemia. J Immunother Cancer. 2021;9(4):e002056. doi:10.1136/jitc-2020-002056
  35. Kymriah. Prescribing information. Novartis Pharmaceuticals Corporation; 2022. https://www.fda.gov/media/107296/download
  36. Tecartus. Prescribing information. Kite Pharma, Inc; 2021. https://www.fda.gov/media/140409/download
  37. Mikhael J, Fowler J, Shah N. Chimeric antigen receptor T-cell therapies: barriers and solutions to access. JCO Oncol Pract. 2022;18(12):800-807. doi:10.1200/OP.22.00315
  38. Teachey DT, O’Connor D. How I treat newly diagnosed T-cell acute lymphoblastic leukemia and T-cell lymphoblastic lymphoma in children. Blood. 2020;135(3):159-166. doi:10.1182/blood.2019001557
  39. Summers RJ, Teachey DT. SOHO state of the art updates and next questions: novel approaches to pediatric T-cell ALL and T-lymphoblastic lymphoma. Clin Lymphoma Myeloma Leuk. 2022;22(10):718-725. doi:10.1016/j.clml.2022.07.010
  40. Hogan LE, Bhatla T, Teachey DT, et al. Efficacy and safety of daratumumab (DARA) in pediatric and young adult patients (pts) with relapsed/refractory T-cell acute lymphoblastic leukemia (ALL) or lymphoblastic lymphoma (LL): results from the phase 2 DELPHINUS study. J Clin Oncol. 2022;40(16 suppl):Abstract 10001. doi:10.1200/JCO.2022.40.16_suppl.10001
  41. Essig S, Li Q, Chen Y, et al. Risk of late effects of treatment in children newly diagnosed with standard-risk acute lymphoblastic leukaemia: a report from the Childhood Cancer Survivor Study. Lancet Oncol. 2014;15(8):841-851. doi:10.1016/S1470-2045(14)70265-7
  42. Long-term follow-up guidelines for survivors of childhood, adolescent, and young adult cancers. Version 5.0. Children’s Oncology Group. October 2018. Accessed April 10, 2023. http://www.survivorshipguidelines.org
  43. Fardell JE, Vetsch J, Trahair T, et al. Health-related quality of life of children on treatment for acute lymphoblastic leukemia: a systematic review. Pediatr Blood Cancer. 2017;64(9). doi:10.1002/pbc.26489
  44. Chantziara S, Musoro J, Rowsell AC, et al; European Organisation for Research and Treatment of Cancer (EORTC) Quality of Life (QLG) and Children’s Leukemia Group (CLG). Quality of life of long-term childhood acute lymphoblastic leukemia survivors: comparison with healthy controls. Psychooncology. 2022;31(12):2159-2168. doi:10.1002/pon.6060
Author and Disclosure Information

Susan Colace, MD, MSCI
Associate Professor of Pediatrics, The Ohio State University School of Medicine;
Co-Director for the Program of Personalized Medicine and Pharmacogenomics in
Hematology/Oncology/BMT, Nationwide Children’s Hospital
Columbus, OH

Susan Colace, MD, MSCI, has disclosed no relevant financial relationships.

Publications
Topics
Author and Disclosure Information

Susan Colace, MD, MSCI
Associate Professor of Pediatrics, The Ohio State University School of Medicine;
Co-Director for the Program of Personalized Medicine and Pharmacogenomics in
Hematology/Oncology/BMT, Nationwide Children’s Hospital
Columbus, OH

Susan Colace, MD, MSCI, has disclosed no relevant financial relationships.

Author and Disclosure Information

Susan Colace, MD, MSCI
Associate Professor of Pediatrics, The Ohio State University School of Medicine;
Co-Director for the Program of Personalized Medicine and Pharmacogenomics in
Hematology/Oncology/BMT, Nationwide Children’s Hospital
Columbus, OH

Susan Colace, MD, MSCI, has disclosed no relevant financial relationships.

Susan Colace, MD, MSCI
Acute lymphocytic leukemia (ALL) is a heterogeneous malignancy that may develop from B or T lymphocytes (B-ALL, T-ALL) and affects patients of all ages.1 In the United States, an estimated 6,540 new cases are diagnosed each year—including 3,100 in individuals aged < 20 years—and approximately 1,390 deaths annually.2,3 It is the most encountered cancer in patients aged < 20 years, and generally carries a good prognosis; almost all younger patients achieve remission with current therapies. Five-year overall survival (OS) is 90% in patients aged < 15 years, 75% in patients aged 15-19 years, and 61% in adolescent/young adult patients (which generally includes patients up to age 39).2,4,5 In contrast, only about 30% of adults with ALL achieve remission with current therapies.


Incidence peaks in children aged 1-4 years, decreasing thereafter. Cases are highest among Native American/Alaskan Native and Hispanic children, and higher in White than Black children.4 ALL is seen more in patients with certain inherited conditions, including Down syndrome, ataxia telangiectasia, neurofibromatosis type 1, and Bloom syndrome.1

Treatment advances have improved remission rates and outcomes for patients. However, relapse is still a leading cause of death for patients of all ages.6 Prompt diagnosis and care are important to optimize outcomes, as treatment delay is associated with poorer survival.7

Pathophysiology

In ALL, abnormal, immature lymphocytes and progenitor B cells/T cells proliferate uncontrollably and eventually replace healthy cells in bone marrow and the lymphatic system. The loss of healthy cells leads to classic symptoms of cytopenia, splenomegaly, and hepatomegaly.1 B cells and T cells are descended from lymphoid stem cells (and are transformed by germline or somatic mutation into pathogenic cells, leading to symptom development and bone marrow dysfunction. Most pediatric patients have extensive bone marrow involvement at diagnosis, with > 25% blast cells in marrow (defined as M3 disease).4

Presentation

Patients usually present with signs and symptoms that are related to disease-associated anemia, thrombocytopenia, or neutropenia; these signs and symptoms may include fatigue or weakness, pale skin, bleeding or bruising easily, fever or infection, joint or extremity pain, B-cell symptoms such as night sweats or unintentional weight loss, and splenomegaly or hepatomegaly. Central nervous system (CNS) symptoms can include stroke-like symptoms due to leukemic cell invasion of CNS vasculature or neuropathies related to increased intracranial pressure. Sometimes, children may present with no symptoms other than joint or extremity pain.1,3,8

Classification

ALL is classified by whether it derives from B-cell or T-cell progenitor cells and, within these, by typical genetic alterations (Table 1).3,9-15 Some cytogenetics are associated with risk assessment as well. Well-identified B-ALL subtypes include Philadelphia (Ph) chromosome-positive, hyper- and hypodiploidy, and KMT2A rearranged, while newer classifications include Ph-like ALL and B-lymphoblastic leukemia with iAMP21. Provisional T-ALL subtypes include early T-cell precursor lymphoblastic leukemia and natural killer cell lymphoblastic leukemia.3

Table 1. Common Genetic Alterations in ALL

B-cell lineage is present in 88% of pediatric and 75%-80% of adult disease. T-ALL is found in about 12% of pediatric patients and 25% of adults.3,8 Familial syndromes associated with ALL are present in about 4% of pediatric patients, including autosomal dominant germline mutations in RUNX1 (T-cell ALL), ETV6 (B-ALL), PAX5 (B-ALL), IKZF1 (B-ALL and T-ALL), and TP53 (low-hypodiploid ALL).3 If a known-familial genotype is identified, families should be referred for genetic counseling and further testing if needed. If germline mutation is suspected, early identification is important; hereditary ALL can influence treatment choice and use of allogeneic transplantation or radiation.3

A third classification crucial to guiding treatment is Ph-positive vs Ph-negative or Ph-like, the latter strongly associated with abnormal B-cell development due to deletions in related genes.3,16 About 3% to 5% of pediatric patients and 25% of adults have Ph-positive ALL.17 The remission failure rate among pediatric patients treated with chemotherapy was 11% in one study, vs 2%-3% among patients with Ph-negative ALL.10

Diagnosis and Risk Stratification

Diagnosis is based on presentation and molecular features, requiring demonstration of ≥ 20% lymphoblasts in bone marrow biopsy or aspirate or ≥ 1,000 circulating lymphoblasts/mL in peripheral blood. Testing can include immunophenotyping using flow cytometry, molecular characterization of baseline leukemic clone, morphology using hematoxylin and eosin staining and Wright/Giemsa staining, and karyotyping.1,3 CNS involvement is assessed using a lumbar spinal tap.1

Risk stratification is based on molecular features (eg, high- and low-risk mutations, Table 1),3,9-15 which are assessed using fluorescence in-situ hybridization, broad-panel next-generation sequencing, and reverse-transcriptase polymerase chain reaction of bone marrow or peripheral blood.Other risk factors include age, CNS involvement, white blood cell (WBC) count, and response to initial induction or consolidation therapy.3

Pediatric patients are assigned standard or high risk based on factors identified by the Children’s Oncology Group and National Comprehensive Cancer Network (NCCN). Patients
aged 1 to < 10 years with WBC < 50 × 109/L are considered standard risk, and all others are considered high risk. Patients with ALL before age 1 have very high risk. All pediatric patients with T-ALL are considered high risk.3 Ph-positive, Ph-like, hypoploidy, failure to achieve remission with induction, and extramedullary disease are high-risk factors as well, whereas hyperploidy and certain mutations convey low risk.3

Newer treatment strategies for initial ALL diagnosis include targeted therapies. One goal of targeted therapy is avoidance of long-term toxicity, leading to improved survival outcomes. Well-studied targeted therapies include the tyrosine kinase inhibitors used in first-line and subsequent treatment of Ph-positive ALL.3

Treatment Options in Relapsed/Refractory ALL

The initial treatment goal is complete remission (CR) defined as minimal residual disease (MRD) < 0.01% on flow cytometry (Table 2).3 Prognosis is dependent on time and location of relapse. Early relapse (< 18 months from diagnosis) predicts poor survival. Relapse in bone marrow is associated with poorer prognosis than relapse in CNS.11-18 Where possible, consolidation with allogeneic hematopoietic cell transplantation improves survival for patients with early relapse.6 Three approaches have advanced treatment options for relapsed/refractory (R/R) B-ALL, all based around common cell markers seen in B-ALL.

Table 2. Response Criteria in ALL

The CD22-directed antibody-drug conjugate inotuzumab ozogamicin is approved for adults with R/R B-ALL. In clinical trials, a higher percentage of patients had results below the MRD threshold, and longer progression-free survival and OS compared with standard care.19,20

Blinatumomab is a bispecific T-cell engager that binds to CD19 on the surface of B-ALL cells and to CD3 on T cells to trigger apoptosis.21 It was first approved for R/R ALL in adults or children, and is also now approved for treatment in remission with MRD ≥ 0.1%. Patients must demonstrate CD19-positive disease to qualify.15-22 For R/R ALL, blinatumomab improves OS and CR rates compared with standard chemotherapy.23

The use of CAR T-cell therapies has expanded greatly with increasing knowledge about their efficacy and safety. In R/R ALL, tisagenlecleucel (tisa-gen) is approved for treatment of patients aged ≤ 25 years, and brexucabtagene autoleucel (brexucel) is approved for treatment of adults.3,24,25 Patients undergoing the CAR T-cell process have apheresis to collect T cells, which are then manufactured before being reinfused into the patient. Depending on local capabilities, the time between T-cell harvest and reinfusion can extend to weeks.3,26,27 Cytoreduction with CAR T-cell therapy can allow previously ineligible patients (due to bulky disease) to undergo transplant. Patients treated in key clinical trials with tisa-gen or brexu-cel achieved high overall remission rates and improved event-free survival and OS rates compared with historical experience.25,28,29 Important toxicities with CAR T-cell therapy are cytokine release syndrome (CRS) and neurotoxicity, which can develop rapidly. NCCN recommends hospitalizing patients at the first sign of either adverse event. Patients can be managed with tocilizumab or steroids for low-grade CRS or steroids for neurotoxicity. The Society for Immunotherapy of Cancer, American Society of Clinical Oncology, and NCCN have guidelines on management of toxicities related to CAR T-cell therapy as well as management of symptoms and other adverse effects of CRS.5,23,24

Programs also incorporate telemedicine for symptom monitoring and follow-up.32-34 Centers providing CAR T-cell therapy must have a certified Risk Evaluation and Mitigation Strategy (REMS), which ensures adherence to specific guidelines for administration, adverse event management, and patient education.35,36 Overcoming technical, social, and financial barriers to CAR T-cell therapy is an ongoing challenge of great interest.37

R/R T-Cell Precursor ALL

Patients with R/R T-ALL have poor prognosis, partly due to limited treatment options. Nelarabine, a nucleoside analog, is the only approved treatment for R/R T-ALL, but has increasingly been used in first-line therapy added to multiagent chemotherapy as a consolidation and maintenance approach to pediatric disease.3,38,39 Four-year DSF in pediatric patients with newly diagnosed T-ALL undergoing treatment incorporating nelarabine was 88.9%.39 Treatment is associated with grade ≥ 3 neurotoxicity in > 10% of patients, and can include CNS toxicity as well as neuropathy.3

In a recently completed phase 2 trial (NCT03384654), daratumumab was added to standard chemotherapy (vincristine, prednisone, PEG-asparaginase, doxorubicin) for R/R T-ALL in pediatric (ages 1-17 years) and young adult patients (age ≥ 18 years).40 Among 24 pediatric patients, CR was 41.7% and overall response rate (ORR; ORR = CR + CRi) was 83% after 1 cycle of treatment. Ten (41.7%) pediatric patients achieved MRD-negative status as well. ORR was 60% in the 5 older patients. All pediatric patients had at least 1 grade ≥ 3 toxicity, but none of the adverse events led to discontinuation.40

Success in achieving MRD-negative responses in patients treated for R/R ALL has increased interest in using targeted therapies for newly diagnosed patients. Recommended treatment approaches are summarized in Table 3.3

Table 3. Recommended Therapy for R/R ALL

Long-Term Follow-Up and Survivorship

A study of > 500 pediatric patients followed for an average 23 years reassuringly found low prevalence of adverse outcomes related to disease or treatment. Major adverse outcomes such as death due to late relapse; secondary malignancy; or development of osteoporosis, cataracts, and diminished functional status were infrequent.41 Most prevalent were growth effects (short stature or growth hormone insufficiency), likely related to certain treatment approaches.41 Guidelines for long-term follow-up of pediatric patients are available from the Children’s Oncology Group.42

A 2017 systematic review concluded that the quality of life for survivors is diminished upon treatment, and persistently over time for some patients.43 In contrast, a 2022 comparison of long-term survivors (median 20.5 years since diagnosis) of pediatric ALL with healthy controls found that survivors had better quality of life in some domains, including general health, vitality, and mental health.44 Smaller percentages of survivors rated themselves happiest about sleep quality, absence of pain, and physical abilities.44

As therapy patterns and options evolve, continued follow-up is important to ensure patients derive optimal benefit from treatment and post-treatment life.

Click to read more from 2023 Rare Diseases Report: Cancers

Susan Colace, MD, MSCI
Acute lymphocytic leukemia (ALL) is a heterogeneous malignancy that may develop from B or T lymphocytes (B-ALL, T-ALL) and affects patients of all ages.1 In the United States, an estimated 6,540 new cases are diagnosed each year—including 3,100 in individuals aged < 20 years—and approximately 1,390 deaths annually.2,3 It is the most encountered cancer in patients aged < 20 years, and generally carries a good prognosis; almost all younger patients achieve remission with current therapies. Five-year overall survival (OS) is 90% in patients aged < 15 years, 75% in patients aged 15-19 years, and 61% in adolescent/young adult patients (which generally includes patients up to age 39).2,4,5 In contrast, only about 30% of adults with ALL achieve remission with current therapies.


Incidence peaks in children aged 1-4 years, decreasing thereafter. Cases are highest among Native American/Alaskan Native and Hispanic children, and higher in White than Black children.4 ALL is seen more in patients with certain inherited conditions, including Down syndrome, ataxia telangiectasia, neurofibromatosis type 1, and Bloom syndrome.1

Treatment advances have improved remission rates and outcomes for patients. However, relapse is still a leading cause of death for patients of all ages.6 Prompt diagnosis and care are important to optimize outcomes, as treatment delay is associated with poorer survival.7

Pathophysiology

In ALL, abnormal, immature lymphocytes and progenitor B cells/T cells proliferate uncontrollably and eventually replace healthy cells in bone marrow and the lymphatic system. The loss of healthy cells leads to classic symptoms of cytopenia, splenomegaly, and hepatomegaly.1 B cells and T cells are descended from lymphoid stem cells (and are transformed by germline or somatic mutation into pathogenic cells, leading to symptom development and bone marrow dysfunction. Most pediatric patients have extensive bone marrow involvement at diagnosis, with > 25% blast cells in marrow (defined as M3 disease).4

Presentation

Patients usually present with signs and symptoms that are related to disease-associated anemia, thrombocytopenia, or neutropenia; these signs and symptoms may include fatigue or weakness, pale skin, bleeding or bruising easily, fever or infection, joint or extremity pain, B-cell symptoms such as night sweats or unintentional weight loss, and splenomegaly or hepatomegaly. Central nervous system (CNS) symptoms can include stroke-like symptoms due to leukemic cell invasion of CNS vasculature or neuropathies related to increased intracranial pressure. Sometimes, children may present with no symptoms other than joint or extremity pain.1,3,8

Classification

ALL is classified by whether it derives from B-cell or T-cell progenitor cells and, within these, by typical genetic alterations (Table 1).3,9-15 Some cytogenetics are associated with risk assessment as well. Well-identified B-ALL subtypes include Philadelphia (Ph) chromosome-positive, hyper- and hypodiploidy, and KMT2A rearranged, while newer classifications include Ph-like ALL and B-lymphoblastic leukemia with iAMP21. Provisional T-ALL subtypes include early T-cell precursor lymphoblastic leukemia and natural killer cell lymphoblastic leukemia.3

Table 1. Common Genetic Alterations in ALL

B-cell lineage is present in 88% of pediatric and 75%-80% of adult disease. T-ALL is found in about 12% of pediatric patients and 25% of adults.3,8 Familial syndromes associated with ALL are present in about 4% of pediatric patients, including autosomal dominant germline mutations in RUNX1 (T-cell ALL), ETV6 (B-ALL), PAX5 (B-ALL), IKZF1 (B-ALL and T-ALL), and TP53 (low-hypodiploid ALL).3 If a known-familial genotype is identified, families should be referred for genetic counseling and further testing if needed. If germline mutation is suspected, early identification is important; hereditary ALL can influence treatment choice and use of allogeneic transplantation or radiation.3

A third classification crucial to guiding treatment is Ph-positive vs Ph-negative or Ph-like, the latter strongly associated with abnormal B-cell development due to deletions in related genes.3,16 About 3% to 5% of pediatric patients and 25% of adults have Ph-positive ALL.17 The remission failure rate among pediatric patients treated with chemotherapy was 11% in one study, vs 2%-3% among patients with Ph-negative ALL.10

Diagnosis and Risk Stratification

Diagnosis is based on presentation and molecular features, requiring demonstration of ≥ 20% lymphoblasts in bone marrow biopsy or aspirate or ≥ 1,000 circulating lymphoblasts/mL in peripheral blood. Testing can include immunophenotyping using flow cytometry, molecular characterization of baseline leukemic clone, morphology using hematoxylin and eosin staining and Wright/Giemsa staining, and karyotyping.1,3 CNS involvement is assessed using a lumbar spinal tap.1

Risk stratification is based on molecular features (eg, high- and low-risk mutations, Table 1),3,9-15 which are assessed using fluorescence in-situ hybridization, broad-panel next-generation sequencing, and reverse-transcriptase polymerase chain reaction of bone marrow or peripheral blood.Other risk factors include age, CNS involvement, white blood cell (WBC) count, and response to initial induction or consolidation therapy.3

Pediatric patients are assigned standard or high risk based on factors identified by the Children’s Oncology Group and National Comprehensive Cancer Network (NCCN). Patients
aged 1 to < 10 years with WBC < 50 × 109/L are considered standard risk, and all others are considered high risk. Patients with ALL before age 1 have very high risk. All pediatric patients with T-ALL are considered high risk.3 Ph-positive, Ph-like, hypoploidy, failure to achieve remission with induction, and extramedullary disease are high-risk factors as well, whereas hyperploidy and certain mutations convey low risk.3

Newer treatment strategies for initial ALL diagnosis include targeted therapies. One goal of targeted therapy is avoidance of long-term toxicity, leading to improved survival outcomes. Well-studied targeted therapies include the tyrosine kinase inhibitors used in first-line and subsequent treatment of Ph-positive ALL.3

Treatment Options in Relapsed/Refractory ALL

The initial treatment goal is complete remission (CR) defined as minimal residual disease (MRD) < 0.01% on flow cytometry (Table 2).3 Prognosis is dependent on time and location of relapse. Early relapse (< 18 months from diagnosis) predicts poor survival. Relapse in bone marrow is associated with poorer prognosis than relapse in CNS.11-18 Where possible, consolidation with allogeneic hematopoietic cell transplantation improves survival for patients with early relapse.6 Three approaches have advanced treatment options for relapsed/refractory (R/R) B-ALL, all based around common cell markers seen in B-ALL.

Table 2. Response Criteria in ALL

The CD22-directed antibody-drug conjugate inotuzumab ozogamicin is approved for adults with R/R B-ALL. In clinical trials, a higher percentage of patients had results below the MRD threshold, and longer progression-free survival and OS compared with standard care.19,20

Blinatumomab is a bispecific T-cell engager that binds to CD19 on the surface of B-ALL cells and to CD3 on T cells to trigger apoptosis.21 It was first approved for R/R ALL in adults or children, and is also now approved for treatment in remission with MRD ≥ 0.1%. Patients must demonstrate CD19-positive disease to qualify.15-22 For R/R ALL, blinatumomab improves OS and CR rates compared with standard chemotherapy.23

The use of CAR T-cell therapies has expanded greatly with increasing knowledge about their efficacy and safety. In R/R ALL, tisagenlecleucel (tisa-gen) is approved for treatment of patients aged ≤ 25 years, and brexucabtagene autoleucel (brexucel) is approved for treatment of adults.3,24,25 Patients undergoing the CAR T-cell process have apheresis to collect T cells, which are then manufactured before being reinfused into the patient. Depending on local capabilities, the time between T-cell harvest and reinfusion can extend to weeks.3,26,27 Cytoreduction with CAR T-cell therapy can allow previously ineligible patients (due to bulky disease) to undergo transplant. Patients treated in key clinical trials with tisa-gen or brexu-cel achieved high overall remission rates and improved event-free survival and OS rates compared with historical experience.25,28,29 Important toxicities with CAR T-cell therapy are cytokine release syndrome (CRS) and neurotoxicity, which can develop rapidly. NCCN recommends hospitalizing patients at the first sign of either adverse event. Patients can be managed with tocilizumab or steroids for low-grade CRS or steroids for neurotoxicity. The Society for Immunotherapy of Cancer, American Society of Clinical Oncology, and NCCN have guidelines on management of toxicities related to CAR T-cell therapy as well as management of symptoms and other adverse effects of CRS.5,23,24

Programs also incorporate telemedicine for symptom monitoring and follow-up.32-34 Centers providing CAR T-cell therapy must have a certified Risk Evaluation and Mitigation Strategy (REMS), which ensures adherence to specific guidelines for administration, adverse event management, and patient education.35,36 Overcoming technical, social, and financial barriers to CAR T-cell therapy is an ongoing challenge of great interest.37

R/R T-Cell Precursor ALL

Patients with R/R T-ALL have poor prognosis, partly due to limited treatment options. Nelarabine, a nucleoside analog, is the only approved treatment for R/R T-ALL, but has increasingly been used in first-line therapy added to multiagent chemotherapy as a consolidation and maintenance approach to pediatric disease.3,38,39 Four-year DSF in pediatric patients with newly diagnosed T-ALL undergoing treatment incorporating nelarabine was 88.9%.39 Treatment is associated with grade ≥ 3 neurotoxicity in > 10% of patients, and can include CNS toxicity as well as neuropathy.3

In a recently completed phase 2 trial (NCT03384654), daratumumab was added to standard chemotherapy (vincristine, prednisone, PEG-asparaginase, doxorubicin) for R/R T-ALL in pediatric (ages 1-17 years) and young adult patients (age ≥ 18 years).40 Among 24 pediatric patients, CR was 41.7% and overall response rate (ORR; ORR = CR + CRi) was 83% after 1 cycle of treatment. Ten (41.7%) pediatric patients achieved MRD-negative status as well. ORR was 60% in the 5 older patients. All pediatric patients had at least 1 grade ≥ 3 toxicity, but none of the adverse events led to discontinuation.40

Success in achieving MRD-negative responses in patients treated for R/R ALL has increased interest in using targeted therapies for newly diagnosed patients. Recommended treatment approaches are summarized in Table 3.3

Table 3. Recommended Therapy for R/R ALL

Long-Term Follow-Up and Survivorship

A study of > 500 pediatric patients followed for an average 23 years reassuringly found low prevalence of adverse outcomes related to disease or treatment. Major adverse outcomes such as death due to late relapse; secondary malignancy; or development of osteoporosis, cataracts, and diminished functional status were infrequent.41 Most prevalent were growth effects (short stature or growth hormone insufficiency), likely related to certain treatment approaches.41 Guidelines for long-term follow-up of pediatric patients are available from the Children’s Oncology Group.42

A 2017 systematic review concluded that the quality of life for survivors is diminished upon treatment, and persistently over time for some patients.43 In contrast, a 2022 comparison of long-term survivors (median 20.5 years since diagnosis) of pediatric ALL with healthy controls found that survivors had better quality of life in some domains, including general health, vitality, and mental health.44 Smaller percentages of survivors rated themselves happiest about sleep quality, absence of pain, and physical abilities.44

As therapy patterns and options evolve, continued follow-up is important to ensure patients derive optimal benefit from treatment and post-treatment life.

Click to read more from 2023 Rare Diseases Report: Cancers

References
  1. Puckett Y, Chan O. Acute lymphocytic leukemia. StatPearls [Internet]. Treasure Island, FL: StatPearls Publishing; 2022. Updated June 27, 2022. Accessed April 10, 2023. https://www.ncbi.nlm.nih.gov/books/NBK459149/
  2. Cancer facts & figures 2023. American Cancer Society. 2023. Accessed April 10, 2023. https://www.cancer.org/content/dam/cancer-org/research/cancer-facts-and-statistics/annual-cancer-facts-and-figures/2023/2023-cancer-facts-and-figures.pdf
  3. National Comprehensive Cancer Network. NCCN clinical practice guidelines in oncology: acute lymphoblastic leukemia. Version 1.2022. April 4, 2022. Accessed April 10, 2023. https://www.nccn.org/professionals/physician_gls/pdf/all.pdf
  4. Childhood acute lymphoblastic leukemia (PDQ)—Health Professional Version. National Cancer Institute. Updated February 16, 2023. Accessed April 10, 2023. https://www.cancer.gov/types/leukemia/hp/child-all-treatment-pdq
  5. National Comprehensive Cancer Network. NCCN clinical practice guidelines in oncology: management of immunotherapy-related toxicities. Version 1.2023. March 10, 2023. Accessed April 10, 2023. https://www.nccn.org/professionals/physician_gls/pdf/immunotherapy.pdf
  6. DuVall AS, Sheade J, Anderson D, et al. Updates in the management of relapsed and refractory acute lymphoplastic leukemia: an urgent plea for new treatments is being answered! JCO Oncol Pract. 2022;18(7):479-487. doi:10.1200/OP.21.00843
  7. Baker JM, To T, Beyene J, Zagorski B, Greenberg ML, Sung L. Influence of length of time to diagnosis and treatment on the survival of children with acute lymphoblastic leukemia: a population-based study. Leuk Res. 2014;38(2):204-209. doi:10.1016/j.leukres.2013.11.014
  8. Acute adult lymphoblastic leukemia (PDQ)—Health Professional Version. National Cancer Institute. Updated February 24, 2023. Accessed April 10, 2023. https://www.cancer.gov/types/leukemia/hp/adult-all-treatment-pdq
  9. Trinquand A, Tanguy-Schmidt A, Ben Abdelali R, et al. Toward a NOTCH1/FBXW7/RAS/PTEN–based oncogenetic risk classification of adult T-cell acute lymphoblastic leukemia: a Group for Research in Adult Acute Lymphoblastic Leukemia Study. J Clin Oncol. 2013;31(34):4333-4342. doi:10.1200/JCO.2012.48.5292
  10. Callens C, Baleydier F, Lengline E, et al. Clinical impact of NOTCH1 and/or FBXW7 mutations, FLASH deletion, and TCR status in pediatric T-cell lymphoblastic lymphoma. J Clin Oncol. 2012;30(16):1966-1973. doi:10.1200/JCO.2011.39.7661
  11. Gao C, Liu SG, Zhang RD, et al. NOTCH1 mutations are associated with favourable long-term prognosis in paediatric T-cell acute lymphoblastic leukaemia: a retrospective study of patients treated on BCH-2003 and CCLG-2008 protocol in China. Br J Haematol. 2014;166(2):221-228. doi:10.1111/bjh.12866
  12. Yang YL, Hsiao CC, Chen HY, et al. Absence of biallelic TCRγ deletion predicts induction failure and poorer outcomes in childhood T-cell acute lymphoblastic leukemia. Pediatr Blood Cancer. 2012;58(6):846-851. doi:10.1002/pbc.24021
  13. Gutierrez A, Dahlberg SE, Neuberg DS, et al. Absence of biallelic TCRgamma deletion predicts early treatment failure in pediatric T-cell acute lymphoblastic leukemia. J Clin Oncol. 2010;28(24):3816-3823. doi:10.1200/JCO.2010.28.3390
  14. Bandapalli OR, Zimmermann M, Kox C, et al. NOTCH1 activation clinically antagonizes the unfavorable effect of PTEN inactivation in BFM-treated children with precursor T-cell acute lymphoblastic leukemia. Haematologica. 2013;98(6):928-936. doi:10.3324/haematol.2012.073585
  15. Palmi C, Savino AM, Silvestri D, et al. CRLF2 over-expression is a poor prognostic marker in children with high risk T-cell acute lymphoblastic leukemia. Oncotarget. 2016;7(37):59260-59272. doi:10.18632/oncotarget.10610
  16. Den Boer ML, van Slegtenhorst M, De Menezes RX, et al. A subtype of childhood acute lymphoblastic leukaemia with poor treatment outcome: a genome-wide classification study. Lancet Oncol. 2009;10(2):125-134. doi:10.1016/S1470-2045(08)70339-5
  17. Aricò M, Schrappe M, Hunger SP, et al. Clinical outcome of children with newly diagnosed Philadelphia chromosome-positive acute lymphoblastic leukemia treated between 1995 and 2005. J Clin Oncol. 2010;28(31):4755-4761. doi:10.1200/JCO.2010.30.1325
  18. Nguyen K, Devidas M, Cheng SC, et al.; Children’s Oncology Group. Factors influencing survival after relapse from acute lymphoblastic leukemia: a Children’s Oncology Group study. Leukemia. 2008;22(12):2142-2150. doi:10.1038/leu.2008.251
  19. Besponsa. Prescribing information. Wyeth Pharmaceuticals Inc; 2017. BESPONSA® (inotuzumab ozogamicin) Dosing & Administration |Safety Info (pfizerpro.com)
  20.  Kantarjian HM, DeAngelo DJ, Stelljes M, et al. Inotuzumab ozogamicin versus standard therapy for acute lymphoblastic leukemia. N Engl J Med. 2016;375(8):740-753. doi:10.1056/NEJMoa1509277
  21. Lv M, Liu Y, Liu W, Xing Y, Zhang S. Immunotherapy for pediatric acute lymphoblastic leukemia: recent advances and future perspectives. Front Immunol. 2022;13:921894. doi:10.3389/fimmu.2022.921894
  22. Blincyto. Prescribing information. Amgen; 2022. https://www.pi.amgen.com/-/media/Project/Amgen/Repository/pi-amgen-com/Blincyto/blincyto_pi_hcp_english.pdf
  23. Kantarjian H, Stein A, Gökbuget N, et al. Blinatumomab versus chemotherapy for advanced acute lymphoblastic leukemia. N Engl J Med. 2017;376(9):836-847. doi:10.1056/NEJMoa1609783
  24. Maude SL, Laetsch TW, Buechner J, et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 2018;378(5):439-448. doi:10.1056/NEJMoa1709866
  25. Shah BD, Ghobadi A, Oluwole OO, et al. KTE-X19 for relapsed or refractory adult B-cell acute lymphoblastic leukaemia: phase 2 results of the single-arm, open-label, multicentre ZUMA-3 study. Lancet. 2021;398(10299):491-502. doi:10.1016/S0140-6736(21)01222-8
  26. Bhaskar ST, Dholaria BR, Singsayadeth S, Savani BN, Oluwole OO. Role of bridging therapy during chimeric antigen receptor T cell therapy. EJHaem. 2021;3(suppl 1):39-45. doi:10.1002/jha2.335
  27. Granroth G, Rosenthal A, McCallen M, et al. Supportive care for patients with lymphoma
    undergoing CAR-T-cell therapy: the advanced practice provider’s perspective. Curr Oncol Rep. 2022;24(12):1863-1872. doi:10.1007/s11912-022-01330-z
  28. Laetsch TW, Maude SL, Rives S, et al. Three-year update of tisagenlecleucel in pediatric and young adult patients with relapsed/refractory acute lymphocytic leukemia in the ELIANA trial. J Clin Oncol. 2023;41(9):1664-1669. doi:10.1200/JCO.22.00642
  29. Shah BD, Ghobadi A, Oluwole OO, et al. Two-year follow-up of KTE-X19 in patients with relapsed or refractory adult B-cell acute lymphoblastic leukemia in ZUMA-3 and its contextualization with SCHOLAR-3, an external historical control study. J Hematol Oncol. 2022;15(1):170. doi:10.1186/s13045-022-01379-0
  30. Maus MV, Alexander S, Bishop MR, et al. Society for Immunotherapy of Cancer (SITC) clinical practice guideline on immune effector cell-related adverse events. J Immunother Cancer. 2020;8(2):e001511. doi:10.1136/jitc-2020-001511
  31. Santomasso BD, Nastoupil LJ, Adkins S, et al. Management of immune-related adverse events in patients treated with chimeric antigen receptor T-cell therapy: ASCO Guideline. J Clin Oncol. 2021;39(35):3978-3992. doi:10.1200/JCO.21.01992
  32. Borogovac A, Keruakous A, Bycko M, et al. Safety and feasibility of outpatient chimeric antigen receptor (CAR) T-cell therapy: experience from a tertiary care center. Bone Marrow Transpl. 2022;57(6):1025-1027. doi:10.1038/s41409-022-01664-z
  33. LeBar K, Murawski S, Umayam S, Quinn V. The role of advanced practice providers and telemedicine in reinventing care: the transition of a CAR T-cell transplantation program to the outpatient setting. J Adv Pract Oncol. 2020;11(7):757-763. doi:10.6004/jadpro.2020.11.7.8
  34. Myers GD, Verneris MR, Goy A, Maziarz RT. Perspectives on outpatient administration of CAR-T cell therapy for aggressive B-cell lymphomas and acute lymphoblastic leukemia. J Immunother Cancer. 2021;9(4):e002056. doi:10.1136/jitc-2020-002056
  35. Kymriah. Prescribing information. Novartis Pharmaceuticals Corporation; 2022. https://www.fda.gov/media/107296/download
  36. Tecartus. Prescribing information. Kite Pharma, Inc; 2021. https://www.fda.gov/media/140409/download
  37. Mikhael J, Fowler J, Shah N. Chimeric antigen receptor T-cell therapies: barriers and solutions to access. JCO Oncol Pract. 2022;18(12):800-807. doi:10.1200/OP.22.00315
  38. Teachey DT, O’Connor D. How I treat newly diagnosed T-cell acute lymphoblastic leukemia and T-cell lymphoblastic lymphoma in children. Blood. 2020;135(3):159-166. doi:10.1182/blood.2019001557
  39. Summers RJ, Teachey DT. SOHO state of the art updates and next questions: novel approaches to pediatric T-cell ALL and T-lymphoblastic lymphoma. Clin Lymphoma Myeloma Leuk. 2022;22(10):718-725. doi:10.1016/j.clml.2022.07.010
  40. Hogan LE, Bhatla T, Teachey DT, et al. Efficacy and safety of daratumumab (DARA) in pediatric and young adult patients (pts) with relapsed/refractory T-cell acute lymphoblastic leukemia (ALL) or lymphoblastic lymphoma (LL): results from the phase 2 DELPHINUS study. J Clin Oncol. 2022;40(16 suppl):Abstract 10001. doi:10.1200/JCO.2022.40.16_suppl.10001
  41. Essig S, Li Q, Chen Y, et al. Risk of late effects of treatment in children newly diagnosed with standard-risk acute lymphoblastic leukaemia: a report from the Childhood Cancer Survivor Study. Lancet Oncol. 2014;15(8):841-851. doi:10.1016/S1470-2045(14)70265-7
  42. Long-term follow-up guidelines for survivors of childhood, adolescent, and young adult cancers. Version 5.0. Children’s Oncology Group. October 2018. Accessed April 10, 2023. http://www.survivorshipguidelines.org
  43. Fardell JE, Vetsch J, Trahair T, et al. Health-related quality of life of children on treatment for acute lymphoblastic leukemia: a systematic review. Pediatr Blood Cancer. 2017;64(9). doi:10.1002/pbc.26489
  44. Chantziara S, Musoro J, Rowsell AC, et al; European Organisation for Research and Treatment of Cancer (EORTC) Quality of Life (QLG) and Children’s Leukemia Group (CLG). Quality of life of long-term childhood acute lymphoblastic leukemia survivors: comparison with healthy controls. Psychooncology. 2022;31(12):2159-2168. doi:10.1002/pon.6060
References
  1. Puckett Y, Chan O. Acute lymphocytic leukemia. StatPearls [Internet]. Treasure Island, FL: StatPearls Publishing; 2022. Updated June 27, 2022. Accessed April 10, 2023. https://www.ncbi.nlm.nih.gov/books/NBK459149/
  2. Cancer facts & figures 2023. American Cancer Society. 2023. Accessed April 10, 2023. https://www.cancer.org/content/dam/cancer-org/research/cancer-facts-and-statistics/annual-cancer-facts-and-figures/2023/2023-cancer-facts-and-figures.pdf
  3. National Comprehensive Cancer Network. NCCN clinical practice guidelines in oncology: acute lymphoblastic leukemia. Version 1.2022. April 4, 2022. Accessed April 10, 2023. https://www.nccn.org/professionals/physician_gls/pdf/all.pdf
  4. Childhood acute lymphoblastic leukemia (PDQ)—Health Professional Version. National Cancer Institute. Updated February 16, 2023. Accessed April 10, 2023. https://www.cancer.gov/types/leukemia/hp/child-all-treatment-pdq
  5. National Comprehensive Cancer Network. NCCN clinical practice guidelines in oncology: management of immunotherapy-related toxicities. Version 1.2023. March 10, 2023. Accessed April 10, 2023. https://www.nccn.org/professionals/physician_gls/pdf/immunotherapy.pdf
  6. DuVall AS, Sheade J, Anderson D, et al. Updates in the management of relapsed and refractory acute lymphoplastic leukemia: an urgent plea for new treatments is being answered! JCO Oncol Pract. 2022;18(7):479-487. doi:10.1200/OP.21.00843
  7. Baker JM, To T, Beyene J, Zagorski B, Greenberg ML, Sung L. Influence of length of time to diagnosis and treatment on the survival of children with acute lymphoblastic leukemia: a population-based study. Leuk Res. 2014;38(2):204-209. doi:10.1016/j.leukres.2013.11.014
  8. Acute adult lymphoblastic leukemia (PDQ)—Health Professional Version. National Cancer Institute. Updated February 24, 2023. Accessed April 10, 2023. https://www.cancer.gov/types/leukemia/hp/adult-all-treatment-pdq
  9. Trinquand A, Tanguy-Schmidt A, Ben Abdelali R, et al. Toward a NOTCH1/FBXW7/RAS/PTEN–based oncogenetic risk classification of adult T-cell acute lymphoblastic leukemia: a Group for Research in Adult Acute Lymphoblastic Leukemia Study. J Clin Oncol. 2013;31(34):4333-4342. doi:10.1200/JCO.2012.48.5292
  10. Callens C, Baleydier F, Lengline E, et al. Clinical impact of NOTCH1 and/or FBXW7 mutations, FLASH deletion, and TCR status in pediatric T-cell lymphoblastic lymphoma. J Clin Oncol. 2012;30(16):1966-1973. doi:10.1200/JCO.2011.39.7661
  11. Gao C, Liu SG, Zhang RD, et al. NOTCH1 mutations are associated with favourable long-term prognosis in paediatric T-cell acute lymphoblastic leukaemia: a retrospective study of patients treated on BCH-2003 and CCLG-2008 protocol in China. Br J Haematol. 2014;166(2):221-228. doi:10.1111/bjh.12866
  12. Yang YL, Hsiao CC, Chen HY, et al. Absence of biallelic TCRγ deletion predicts induction failure and poorer outcomes in childhood T-cell acute lymphoblastic leukemia. Pediatr Blood Cancer. 2012;58(6):846-851. doi:10.1002/pbc.24021
  13. Gutierrez A, Dahlberg SE, Neuberg DS, et al. Absence of biallelic TCRgamma deletion predicts early treatment failure in pediatric T-cell acute lymphoblastic leukemia. J Clin Oncol. 2010;28(24):3816-3823. doi:10.1200/JCO.2010.28.3390
  14. Bandapalli OR, Zimmermann M, Kox C, et al. NOTCH1 activation clinically antagonizes the unfavorable effect of PTEN inactivation in BFM-treated children with precursor T-cell acute lymphoblastic leukemia. Haematologica. 2013;98(6):928-936. doi:10.3324/haematol.2012.073585
  15. Palmi C, Savino AM, Silvestri D, et al. CRLF2 over-expression is a poor prognostic marker in children with high risk T-cell acute lymphoblastic leukemia. Oncotarget. 2016;7(37):59260-59272. doi:10.18632/oncotarget.10610
  16. Den Boer ML, van Slegtenhorst M, De Menezes RX, et al. A subtype of childhood acute lymphoblastic leukaemia with poor treatment outcome: a genome-wide classification study. Lancet Oncol. 2009;10(2):125-134. doi:10.1016/S1470-2045(08)70339-5
  17. Aricò M, Schrappe M, Hunger SP, et al. Clinical outcome of children with newly diagnosed Philadelphia chromosome-positive acute lymphoblastic leukemia treated between 1995 and 2005. J Clin Oncol. 2010;28(31):4755-4761. doi:10.1200/JCO.2010.30.1325
  18. Nguyen K, Devidas M, Cheng SC, et al.; Children’s Oncology Group. Factors influencing survival after relapse from acute lymphoblastic leukemia: a Children’s Oncology Group study. Leukemia. 2008;22(12):2142-2150. doi:10.1038/leu.2008.251
  19. Besponsa. Prescribing information. Wyeth Pharmaceuticals Inc; 2017. BESPONSA® (inotuzumab ozogamicin) Dosing & Administration |Safety Info (pfizerpro.com)
  20.  Kantarjian HM, DeAngelo DJ, Stelljes M, et al. Inotuzumab ozogamicin versus standard therapy for acute lymphoblastic leukemia. N Engl J Med. 2016;375(8):740-753. doi:10.1056/NEJMoa1509277
  21. Lv M, Liu Y, Liu W, Xing Y, Zhang S. Immunotherapy for pediatric acute lymphoblastic leukemia: recent advances and future perspectives. Front Immunol. 2022;13:921894. doi:10.3389/fimmu.2022.921894
  22. Blincyto. Prescribing information. Amgen; 2022. https://www.pi.amgen.com/-/media/Project/Amgen/Repository/pi-amgen-com/Blincyto/blincyto_pi_hcp_english.pdf
  23. Kantarjian H, Stein A, Gökbuget N, et al. Blinatumomab versus chemotherapy for advanced acute lymphoblastic leukemia. N Engl J Med. 2017;376(9):836-847. doi:10.1056/NEJMoa1609783
  24. Maude SL, Laetsch TW, Buechner J, et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 2018;378(5):439-448. doi:10.1056/NEJMoa1709866
  25. Shah BD, Ghobadi A, Oluwole OO, et al. KTE-X19 for relapsed or refractory adult B-cell acute lymphoblastic leukaemia: phase 2 results of the single-arm, open-label, multicentre ZUMA-3 study. Lancet. 2021;398(10299):491-502. doi:10.1016/S0140-6736(21)01222-8
  26. Bhaskar ST, Dholaria BR, Singsayadeth S, Savani BN, Oluwole OO. Role of bridging therapy during chimeric antigen receptor T cell therapy. EJHaem. 2021;3(suppl 1):39-45. doi:10.1002/jha2.335
  27. Granroth G, Rosenthal A, McCallen M, et al. Supportive care for patients with lymphoma
    undergoing CAR-T-cell therapy: the advanced practice provider’s perspective. Curr Oncol Rep. 2022;24(12):1863-1872. doi:10.1007/s11912-022-01330-z
  28. Laetsch TW, Maude SL, Rives S, et al. Three-year update of tisagenlecleucel in pediatric and young adult patients with relapsed/refractory acute lymphocytic leukemia in the ELIANA trial. J Clin Oncol. 2023;41(9):1664-1669. doi:10.1200/JCO.22.00642
  29. Shah BD, Ghobadi A, Oluwole OO, et al. Two-year follow-up of KTE-X19 in patients with relapsed or refractory adult B-cell acute lymphoblastic leukemia in ZUMA-3 and its contextualization with SCHOLAR-3, an external historical control study. J Hematol Oncol. 2022;15(1):170. doi:10.1186/s13045-022-01379-0
  30. Maus MV, Alexander S, Bishop MR, et al. Society for Immunotherapy of Cancer (SITC) clinical practice guideline on immune effector cell-related adverse events. J Immunother Cancer. 2020;8(2):e001511. doi:10.1136/jitc-2020-001511
  31. Santomasso BD, Nastoupil LJ, Adkins S, et al. Management of immune-related adverse events in patients treated with chimeric antigen receptor T-cell therapy: ASCO Guideline. J Clin Oncol. 2021;39(35):3978-3992. doi:10.1200/JCO.21.01992
  32. Borogovac A, Keruakous A, Bycko M, et al. Safety and feasibility of outpatient chimeric antigen receptor (CAR) T-cell therapy: experience from a tertiary care center. Bone Marrow Transpl. 2022;57(6):1025-1027. doi:10.1038/s41409-022-01664-z
  33. LeBar K, Murawski S, Umayam S, Quinn V. The role of advanced practice providers and telemedicine in reinventing care: the transition of a CAR T-cell transplantation program to the outpatient setting. J Adv Pract Oncol. 2020;11(7):757-763. doi:10.6004/jadpro.2020.11.7.8
  34. Myers GD, Verneris MR, Goy A, Maziarz RT. Perspectives on outpatient administration of CAR-T cell therapy for aggressive B-cell lymphomas and acute lymphoblastic leukemia. J Immunother Cancer. 2021;9(4):e002056. doi:10.1136/jitc-2020-002056
  35. Kymriah. Prescribing information. Novartis Pharmaceuticals Corporation; 2022. https://www.fda.gov/media/107296/download
  36. Tecartus. Prescribing information. Kite Pharma, Inc; 2021. https://www.fda.gov/media/140409/download
  37. Mikhael J, Fowler J, Shah N. Chimeric antigen receptor T-cell therapies: barriers and solutions to access. JCO Oncol Pract. 2022;18(12):800-807. doi:10.1200/OP.22.00315
  38. Teachey DT, O’Connor D. How I treat newly diagnosed T-cell acute lymphoblastic leukemia and T-cell lymphoblastic lymphoma in children. Blood. 2020;135(3):159-166. doi:10.1182/blood.2019001557
  39. Summers RJ, Teachey DT. SOHO state of the art updates and next questions: novel approaches to pediatric T-cell ALL and T-lymphoblastic lymphoma. Clin Lymphoma Myeloma Leuk. 2022;22(10):718-725. doi:10.1016/j.clml.2022.07.010
  40. Hogan LE, Bhatla T, Teachey DT, et al. Efficacy and safety of daratumumab (DARA) in pediatric and young adult patients (pts) with relapsed/refractory T-cell acute lymphoblastic leukemia (ALL) or lymphoblastic lymphoma (LL): results from the phase 2 DELPHINUS study. J Clin Oncol. 2022;40(16 suppl):Abstract 10001. doi:10.1200/JCO.2022.40.16_suppl.10001
  41. Essig S, Li Q, Chen Y, et al. Risk of late effects of treatment in children newly diagnosed with standard-risk acute lymphoblastic leukaemia: a report from the Childhood Cancer Survivor Study. Lancet Oncol. 2014;15(8):841-851. doi:10.1016/S1470-2045(14)70265-7
  42. Long-term follow-up guidelines for survivors of childhood, adolescent, and young adult cancers. Version 5.0. Children’s Oncology Group. October 2018. Accessed April 10, 2023. http://www.survivorshipguidelines.org
  43. Fardell JE, Vetsch J, Trahair T, et al. Health-related quality of life of children on treatment for acute lymphoblastic leukemia: a systematic review. Pediatr Blood Cancer. 2017;64(9). doi:10.1002/pbc.26489
  44. Chantziara S, Musoro J, Rowsell AC, et al; European Organisation for Research and Treatment of Cancer (EORTC) Quality of Life (QLG) and Children’s Leukemia Group (CLG). Quality of life of long-term childhood acute lymphoblastic leukemia survivors: comparison with healthy controls. Psychooncology. 2022;31(12):2159-2168. doi:10.1002/pon.6060
Publications
Publications
Topics
Article Type
Display Headline
Progress in Management of Advanced Acute Lymphocytic Leukemia in Children
Display Headline
Progress in Management of Advanced Acute Lymphocytic Leukemia in Children
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Thu, 06/22/2023 - 22:00
Un-Gate On Date
Thu, 06/22/2023 - 22:00
Use ProPublica
CFC Schedule Remove Status
Thu, 06/22/2023 - 22:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Treatment Needs of Older Adults With Newly Diagnosed Acute Myeloid Leukemia

Article Type
Changed
Mon, 06/26/2023 - 12:34
Display Headline
Treatment Needs of Older Adults With Newly Diagnosed Acute Myeloid Leukemia

Harry Erba, MD, PhD
Defining “Unfit” for Intensive Chemotherapy

Within the last 40 years, younger fit patients have benefited from intensive chemotherapy regimens for acute myeloid leukemia (AML) with improved survival, and the possibility of long-term disease-free survival (DFS) (“cure”).1 Older patients are often considered too unfit for standard curative treatment with intensive induction chemotherapy followed by consolidation chemotherapy, allogeneic hematopoietic cell transplantation (allo-HCT), or both.2-4 Higher induction mortality and poor overall survival (OS) are associated with worse performance status, organ impairment, significant comorbidities, and declining cognitive function, all of which are more common with advancing age. Although the suggested criteria for determining unfitness have not been validated (Table 1), they can provide guidance in clinical practice.2-5

Table 1. Criteria to Define Unfitness for Intensive Chemotherapy to Treat AML

The National Comprehensive Cancer Network (NCCN) panel recommends the consideration of a patient’s performance status and comorbid conditions in addition to their age to determine a patient’s fitness for intensive induction therapy.6 Adverse disease features should also be considered, because disease biology may make intensive chemotherapy futile or inappropriate. For example, the mutational driver tumor protein p53 (TP53) appears at a higher frequency in older adults than younger adults and is associated with dismal outcomes even with intensive chemotherapy. Likewise, the spliceosome and chromatin modifier gene mutations are more common in older patients with AML and confer a worse OS with intensive therapy.6,7 Older unfit patients faced a difficult decision: proceed with intensive therapy with some possibility of long-term survival but risk of early mortality and significant toxicity, or opt for supportive care and palliative chemotherapy, such as the hypomethylating agents (HMAs) or low-dose cytarabine, with much shorter survival.

Guidelines for Treating Older Unfit Patients

Evidence-based guidelines for managing older adults with newly diagnosed AML were developed by the American Society of Hematology in 2020; however, these guidelines were released prior to the results of several clinical trials involving older patients with AML (Table 2).In 2022, the European LeukemiaNet (ELN) recommendations were updated to include new therapeutic agents that target specific mutations in genes such as tyrosine kinase 3 (FLT3), isocitrate dehydrogenase 1 (IDH1), isocitrate dehydrogenase 2 (IDH2), and B-cell lymphoma 2 (BCL2). Given the important effects of genetic aberrations on disease phenotype, treatment options, and outcomes, screening for genetic aberrations at diagnosis is now essential.9

Table 2. Treatment Landscape for Older Unfit Patients

The potential for clonal evolution leading to new actionable targets that were not present at diagnosis highlights the importance of reevaluation of genetic aberrations throughout clinical progression. Actionable targets can include mutations in IDH1/IDH2, FLT3-internal tandem duplication or FLT3 tyrosine kinase domain.9

Treatment Landscape

Since 2018, several therapeutic agents have been added to the treatment armamentarium that can induce longer-term complete remission (CR) for older unfit patients with newly diagnosed AML (Table 2).

Management of Primary AML With Less Intensive Induction Therapy

VIALE-A established a new standard of care for older unfit patients by demonstrating the benefit of adding the BCL2 inhibitor venetoclax (VEN) to azacitidine (AZA).2 VIALE-A demonstrated that the rate of CR plus CR with partial hematologic recovery (CRi) was 65% for VEN plus AZA and 18% for AZA. Most remissions in the AZA/VEN arm occurred rapidly in the first 2 cycles. The median survival improved from 9.6 months with AZA to 14.7 months with AZA/VEN. An improvement in survival with VEN and low-dose cytarabine also emerged in a 6-month post hoc analysis of the VIALE-C trial.10 Various other trials examining targeted therapies on specific mutations have provided mixed results in the front-line setting.13,14,18 It is important to note that a recent systematic review found that 12% to 25% of patients who were unfit for intensive therapy were successfully bridged to HCT.19

Management of Postremission Response

Patients with a longer duration of first remission have demonstrated better survival outcomes.15 Two trials have examined postremission therapy in the setting of prior intensive therapy. HOVON97 enrolled older patients who achieved CR/CRi after 2 cycles of intensive therapy to receive either AZA postremission or no further treatment. The proportion of patients with DFS at 12 months was greater in the AZA maintenance group than in the observation group (64% vs 42%), but significant DFS improvement did not translate into improved OS.20 QUAZAR AML-001 demonstrated that OS was longer for older patients receiving maintenance therapy with CC-486 (a non-bioequivalent oral formulation of AZA) vs placebo (24.7 vs 14.8 months).15 CC-486 was FDA-approved for maintenance therapy after intensive induction with or without consolidation in patients who are not candidates for allo-HCT. However, limited evidence exists specifically for postremission therapy in unfit patients who have received less intensive therapy. Continuation of the lower intensive therapy is recommended until disease progression.6 No data are available to support the use of oral AZA therapy alone for maintenance of remission following HMA/VEN-induced remissions.

Management of Relapsed and Refractory AML

Nearly 50% of patients with AML experience relapse and up to 40% may be refractory.19 Importantly, patients who were considered fit for intensive therapy may not remain so with relapsed or refractory AML (r/rAML), so patients should be evaluated for fitness for an intensive salvage regimen. Similar to assessing fitness for induction therapy, no standard definition of fitness exists for r/rAML.19

Disease control is the goal for patients with r/rAML who are unfit for intensive salvage therapy; however, treatment options remain limited and prognosis is poor.19 Depending on the patient’s cytogenetic profile, management can include HMA with or without VEN, glasdegib with LDAC, gilteritinib, ivosidenib or enasidenib, or gemtuzumab ozogamicin.9 Only a few studies have been published involving the r/rAML population not eligible for intensive salvage regimen, and guidelines are needed for this population.19 Thus, the ELN recommends that clinical trial enrollment be considered for patients with r/rAML.9

Management of Secondary AML or High-risk AML

Compared with de novo AML, both secondary AML (sAML) and therapy-related AML (tAML) have been associated with inferior outcomes. Factors that influence poor outcomes can include older age, comorbidities, persistent malignant disease or relapse of primary malignancy, treatment-induced depletion of hematopoietic reserves and/or prolonged myelosuppression, and genetic abnormalities, such as TP53 mutations.21

CPX-351 is a dual drug that contains cytarabine and daunorubicin.9,22 An open-label study (NCT01696084) compared CPX-351 with conventional cytarabine and daunorubicin (induction and consolidation therapy) in older patients (aged 60-75 years) with newly diagnosed high-risk/sAML who were considered fit for intensive therapy. The OS for CPX-351 was longer (9.56 vs 5.95 months) and the safety profiles were similar between the treatment groups.23 Patients achieving CR/CRi received up to 2 cycles of consolidation with CPX-351. An exploratory analysis of this subgroup revealed median OS was longer with CPX-351 consolidation (25.43 vs 8.53 months).22 Patients with TP53 mutations had poor treatment outcomes regardless of treatment arm, whereas patients with sAML-type mutations including spliceosome and chromatin modifier genes had longer OS with CPX-351 therapy.24 The 5-year results of this trial indicate that the survival benefit of CPX-351 was maintained.25 However, data from a retrospective review involving 136 patients with either sAML or AML with myelodysplasia-related changes revealed no difference in survival outcomes between patients treated with either HMA/VEN or CPX-351.26

Case Study: Elderly Woman With Newly Diagnosed AML

In 2018, Ms. W, age 69 years, was diagnosed with seropositive, non-erosive rheumatoid arthritis; she began methotrexate 17.5 mg per week split dosing in conjunction with oral folic acid 2 mg/d with varying doses based on symptoms. Her comorbidities included recurrent episodes of diverticulitis, hypertension, hypothyroidism, obstructive sleep apnea, and gastrointestinal reflux disease. On February 4, 2021, her methotrexate was increased to 20 mg and required intermittent prednisone tapers for flares. In November 2021, a blood test revealed she had a decreased white blood cell (WBC) count at 1.8 K/μL, and her methotrexate dose was decreased to 15 mg weekly. Despite the dose reduction, she had grade 3 neutropenia and anemia (WBC: 0.7 K/μL; HGB:10.5 g/dL) with a normal platelet count (PLT: 165,000/μL). Methotrexate was discontinued and leucovorin was initiated. She then had only modest improvement in her lab values and peripheral blood blasts. 

On March 17, 2022, she underwent a bone marrow biopsy and aspirate, which resulted in a diagnosis of AML. She had 55% blasts in a 90% cellular bone marrow with mild reticulin fibrosis and numerous circulating blasts. She was classified as having AML without maturation (FAB AML-M1). Flow cytometry detected a phenotypically abnormal population with CD45 expression and side scatter/forward scatter features of small-to-medium sized blasts, accounting for 23% of total cells. The chromosome analysis demonstrated a normal female karyotype in all 19 available metaphases. Polymerase chain reaction analysis was negative for FLT3-ITD, FLT3-TKD, and NPM1 mutations and positive for an IDH1 R132C missense mutation. The myeloid gene panel identified only a single pathogenic variant, IDH1 R132C (variant allele frequency [VAF] 21.2%), and a variant of unknown significance DNMT3A A575P (VAF 25.7%).

Noting that she does not have favorable risk features, we discussed treatment options. Although she is a candidate for curative therapy, the patient was not interested in pursuing allo-HCT. Her history of diverticulitis is concerning for tolerating intensive chemotherapy. In addition, her immunosuppressive therapy increases her risk for opportunistic infections. Based on the available data from the AGILE and VIALE studies and associated potential adverse reactions, she opted for starting treatment with AZA and IVO.

On March 31, 2022, she began receiving AZA 75 mg/m2 intravenous (IV) once daily days 1-7 and oral IVO 500 mg once daily continuously. She has received 12 cycles and has not needed transfusion. She has not had febrile neutropenia or symptoms of differentiation syndrome. On March 24, 2023, she underwent laparoscopic cholecystectomy, because an ultrasound revealed cholelithiasis, abnormal gallbladder wall thickening, and pericholecystic fluid. She was discharged home the following day and is continuing with AZA/ivosidenib.

Click to read more from 2023 Rare Diseases Report: Cancers

References
  1. Schlenk RF. Acute myeloid leukemia: introduction to a series highlighting progress and ongoing challenges. Haematologica. 2023;108(2):306-307. doi:10.3324/haematol.2022.280803
  2. DiNardo CD, Jonas BA, Pullarkat V, et al. Azacitidine and venetoclax in previously untreated acute myeloid leukemia. N Engl J Med. 2020;383(7):617-629. doi:10.1056/NEJMoa2012971
  3. DiNardo CD, Wei AH. How I treat acute myeloid leukemia in the era of new drugs. Blood. 2020;135(2):85-96. doi:10.1182/blood.2019001239
  4. Huerga-Domínguez S, Villar S, Prósper F, Alfonso-Piérola A. Updates on the management of acute myeloid leukemia. Cancers (Basel). 2022;14(19):4756. doi:10.3390/cancers14194756
  5. Ferrara F, Barosi G, Venditti A, et al. Consensus-based definition of unfitness to intensive and non-intensive chemotherapy in acute myeloid leukemia: a project of SIE, SIES and GITMO group on a new tool for therapy decision making. Leukemia. 2013;27(5):997-999. doi:10.1038/leu.2012.303
  6. Tallman MS, Wang ES, Altman JK, et al. Acute myeloid leukemia, version 3.2019, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2019;17(6):721-749. doi:10.6004/jnccn.2019.0028
  7. Burd A, Levine RL, Ruppert AS, et al. Precision medicine treatment in acute myeloid leukemia using prospective genomic profiling: feasibility and preliminary efficacy of the Beat AML Master Trial. Nat Med. 2020;26(12):1852-1858. doi:10.1038/s41591-020-1089-8
  8. Sekeres MA, Guyatt G, Abel G, et al. American Society of Hematology 2020 guidelines for treating newly diagnosed acute myeloid leukemia in older adults. Blood Adv. 2020;4(15):3528-3549. doi:10.1182/bloodadvances.2020001920
  9. Döhner H, Wei AH, Appelbaum FR, et al. Diagnosis and management of AML in adults: 2022 recommendations from an international expert panel on behalf of the ELN. Blood. 2022;140(12):1345-1377. doi:10.1182/blood.2022016867
  10. Wei AH, Montesinos P, Ivanov V, et al. Venetoclax plus LDAC for newly diagnosed AML ineligible for intensive chemotherapy: a phase 3 randomized placebo-controlled trial. Blood. 2020;135(24):2137-2145. doi:10.1182/blood.2020004856
  11. Amadori S, Suciu S, Selleslag D, et al. Gemtuzumab ozogamicin versus best supportive care in older patients with newly diagnosed acute myeloid leukemia unsuitable for intensive chemotherapy: results of the randomized phase III EORTC-GIMEMA AML-19 trial. J Clin Oncol. 2016;34(9):972-979. doi:10.1200/JCO.2015.64.0060
  12. Cortes JE, Heidel FH, Hellmann A, et al. Randomized comparison of low dose cytarabine with or without glasdegib in patients with newly diagnosed acute myeloid leukemia or high-risk myelodysplastic syndrome. Leukemia. 2019;33(2):379-389. doi:10.1038/s41375-018-0312-9
  13. Montesinos P, Recher C, Vives S, et al. Ivosidenib and azacitidine in IDH1-mutated acute myeloid leukemia. N Engl J Med. 2022;386(16):1519-1531. doi:10.1056/NEJMoa2117344
  14. Wang ES, Montesinos P, Minden MD, et al. Phase 3 trial of gilteritinib plus azacitidine vs azacitidine for newly diagnosed FLT3mut+ AML ineligible for intensive chemotherapy. Blood. 2022;140(17):1845-1857. doi:10.1182/blood.2021014586
  15. Wei AH, Döhner H, Pocock C, et al; QUAZAR AML-001 Trial Investigators. Oral azacitidine maintenance therapy for acute myeloid leukemia in first remission. N Engl J Med. 2020;383(26):2526-2537. doi:10.1056/NEJMoa2004444
  16. Perl AE, Martinelli G, Cortes JE, et al. Gilteritinib or chemotherapy for relapsed or refractory FLT3-mutated AML. N Engl J Med. 2019;381(18):1728-1740. doi:10.1056/NEJMoa1902688
  17. Konopleva MY, Röllig C, Cavenagh J, et al. Idasanutlin plus cytarabine in relapsed or refractory acute myeloid leukemia: results of the MIRROS trial. Blood Adv. 2022;6(14):4147-4156. doi:10.1182/bloodadvances.2021006303
  18. Pollyea DA, DiNardo CD, Arellano ML, et al. Impact of venetoclax and azacitidine in treatment-naïve patients with acute myeloid leukemia and IDH1/2 mutations. Clin Cancer Res. 2022;28(13):2753-2761. doi:10.1158/1078-0432.CCR-21-3467
  19. Russell-Smith TA, Gurskyte L, Muresan B, et al. Efficacy of non-intensive therapies approved for relapsed/refractory acute myeloid leukemia: a systematic literature review. Future Oncol. 2022;18(16):2029-2039. doi:10.2217/fon-2021-1355
  20. Huls G, Chitu DA, Havelange V, et al; Dutch-Belgian Hemato-Oncology Cooperative Group (HOVON). Azacitidine maintenance after intensive chemotherapy improves DFS in older AML patients. Blood. 2019;133(13):1457-1464. doi:10.1182/blood-2018-10-879866
  21. Granfeldt Østgård LS, Medeiros BC, Sengeløv H, et al. Epidemiology and clinical significance of secondary and therapy-related acute myeloid leukemia: a national population-based cohort study. J Clin Oncol. 2015;33(31):3641-3649. doi:10.1200/JCO.2014.60.0890
  22. Kolitz JE, Strickland SA, Cortes JE, et al. Consolidation outcomes in CPX-351 versus cytarabine/daunorubicin-treated older patients with high-risk/secondary acute myeloid leukemia. Leuk Lymphoma. 2020;61(3):631-640. doi:10.1080/1042819.2019.1688320
  23. Lancet JE, Uy GL, Cortes JE, et al. CPX-351 (cytarabine and daunorubicin) liposome for injection versus conventional cytarabine plus daunorubicin in older patients with newly diagnosed secondary acute myeloid leukemia. J Clin Oncol. 2018;36(26):2684-2692. doi:10.1200/JCO.2017.77.6112
  24. Lindsley RC, Gibson CJ, Murdock HM, et al. Genetic characteristics and outcomes by mutation status in a phase 3 study of CPX-351 versus 7+3 in older adults with newly diagnosed, high-risk/secondary acute myeloid leukemia (AML). Blood. 2019;134(suppl 1):15. doi:10.1182/blood-2019-124500
  25. Lancet JE, Uy GL, Newell LF, et al. CPX-351 versus 7+3 cytarabine and daunorubicin chemotherapy in older adults with newly diagnosed high-risk or secondary acute myeloid leukaemia: 5-year results of a randomised, open-label, multicentre, phase 3 trial. Lancet Haematol. 2021;8(7):e481-e491. doi:10.1016/S2352-3026(21)00134-4
  26. Alharthy H, Alkaabba F, Williams M, et al. Outcomes of newly diagnosed therapy-related AML and AML with myelodysplasia-related changes treated with 7+3, hypomethylating agents with or without venetoclax and CPX-351: a retrospective cohort study. Blood. 2022;140(suppl 1):9025-9026. doi:10.1182/blood-2022-170688
Author and Disclosure Information

Harry Erba, MD, PhD
Professor, Department of Medicine
Director of Leukemia Program, Division of Hematologic Malignancies and Cellular Therapy
Department of Medicine
Duke University
Durham, NC

Harry Erba, MD, PhD, has disclosed the following relevant financial relationships:
Received income in an amount equal to or greater than $250 from: Astellas; Daiichi Sankyo

Publications
Topics
Author and Disclosure Information

Harry Erba, MD, PhD
Professor, Department of Medicine
Director of Leukemia Program, Division of Hematologic Malignancies and Cellular Therapy
Department of Medicine
Duke University
Durham, NC

Harry Erba, MD, PhD, has disclosed the following relevant financial relationships:
Received income in an amount equal to or greater than $250 from: Astellas; Daiichi Sankyo

Author and Disclosure Information

Harry Erba, MD, PhD
Professor, Department of Medicine
Director of Leukemia Program, Division of Hematologic Malignancies and Cellular Therapy
Department of Medicine
Duke University
Durham, NC

Harry Erba, MD, PhD, has disclosed the following relevant financial relationships:
Received income in an amount equal to or greater than $250 from: Astellas; Daiichi Sankyo

Harry Erba, MD, PhD
Defining “Unfit” for Intensive Chemotherapy

Within the last 40 years, younger fit patients have benefited from intensive chemotherapy regimens for acute myeloid leukemia (AML) with improved survival, and the possibility of long-term disease-free survival (DFS) (“cure”).1 Older patients are often considered too unfit for standard curative treatment with intensive induction chemotherapy followed by consolidation chemotherapy, allogeneic hematopoietic cell transplantation (allo-HCT), or both.2-4 Higher induction mortality and poor overall survival (OS) are associated with worse performance status, organ impairment, significant comorbidities, and declining cognitive function, all of which are more common with advancing age. Although the suggested criteria for determining unfitness have not been validated (Table 1), they can provide guidance in clinical practice.2-5

Table 1. Criteria to Define Unfitness for Intensive Chemotherapy to Treat AML

The National Comprehensive Cancer Network (NCCN) panel recommends the consideration of a patient’s performance status and comorbid conditions in addition to their age to determine a patient’s fitness for intensive induction therapy.6 Adverse disease features should also be considered, because disease biology may make intensive chemotherapy futile or inappropriate. For example, the mutational driver tumor protein p53 (TP53) appears at a higher frequency in older adults than younger adults and is associated with dismal outcomes even with intensive chemotherapy. Likewise, the spliceosome and chromatin modifier gene mutations are more common in older patients with AML and confer a worse OS with intensive therapy.6,7 Older unfit patients faced a difficult decision: proceed with intensive therapy with some possibility of long-term survival but risk of early mortality and significant toxicity, or opt for supportive care and palliative chemotherapy, such as the hypomethylating agents (HMAs) or low-dose cytarabine, with much shorter survival.

Guidelines for Treating Older Unfit Patients

Evidence-based guidelines for managing older adults with newly diagnosed AML were developed by the American Society of Hematology in 2020; however, these guidelines were released prior to the results of several clinical trials involving older patients with AML (Table 2).In 2022, the European LeukemiaNet (ELN) recommendations were updated to include new therapeutic agents that target specific mutations in genes such as tyrosine kinase 3 (FLT3), isocitrate dehydrogenase 1 (IDH1), isocitrate dehydrogenase 2 (IDH2), and B-cell lymphoma 2 (BCL2). Given the important effects of genetic aberrations on disease phenotype, treatment options, and outcomes, screening for genetic aberrations at diagnosis is now essential.9

Table 2. Treatment Landscape for Older Unfit Patients

The potential for clonal evolution leading to new actionable targets that were not present at diagnosis highlights the importance of reevaluation of genetic aberrations throughout clinical progression. Actionable targets can include mutations in IDH1/IDH2, FLT3-internal tandem duplication or FLT3 tyrosine kinase domain.9

Treatment Landscape

Since 2018, several therapeutic agents have been added to the treatment armamentarium that can induce longer-term complete remission (CR) for older unfit patients with newly diagnosed AML (Table 2).

Management of Primary AML With Less Intensive Induction Therapy

VIALE-A established a new standard of care for older unfit patients by demonstrating the benefit of adding the BCL2 inhibitor venetoclax (VEN) to azacitidine (AZA).2 VIALE-A demonstrated that the rate of CR plus CR with partial hematologic recovery (CRi) was 65% for VEN plus AZA and 18% for AZA. Most remissions in the AZA/VEN arm occurred rapidly in the first 2 cycles. The median survival improved from 9.6 months with AZA to 14.7 months with AZA/VEN. An improvement in survival with VEN and low-dose cytarabine also emerged in a 6-month post hoc analysis of the VIALE-C trial.10 Various other trials examining targeted therapies on specific mutations have provided mixed results in the front-line setting.13,14,18 It is important to note that a recent systematic review found that 12% to 25% of patients who were unfit for intensive therapy were successfully bridged to HCT.19

Management of Postremission Response

Patients with a longer duration of first remission have demonstrated better survival outcomes.15 Two trials have examined postremission therapy in the setting of prior intensive therapy. HOVON97 enrolled older patients who achieved CR/CRi after 2 cycles of intensive therapy to receive either AZA postremission or no further treatment. The proportion of patients with DFS at 12 months was greater in the AZA maintenance group than in the observation group (64% vs 42%), but significant DFS improvement did not translate into improved OS.20 QUAZAR AML-001 demonstrated that OS was longer for older patients receiving maintenance therapy with CC-486 (a non-bioequivalent oral formulation of AZA) vs placebo (24.7 vs 14.8 months).15 CC-486 was FDA-approved for maintenance therapy after intensive induction with or without consolidation in patients who are not candidates for allo-HCT. However, limited evidence exists specifically for postremission therapy in unfit patients who have received less intensive therapy. Continuation of the lower intensive therapy is recommended until disease progression.6 No data are available to support the use of oral AZA therapy alone for maintenance of remission following HMA/VEN-induced remissions.

Management of Relapsed and Refractory AML

Nearly 50% of patients with AML experience relapse and up to 40% may be refractory.19 Importantly, patients who were considered fit for intensive therapy may not remain so with relapsed or refractory AML (r/rAML), so patients should be evaluated for fitness for an intensive salvage regimen. Similar to assessing fitness for induction therapy, no standard definition of fitness exists for r/rAML.19

Disease control is the goal for patients with r/rAML who are unfit for intensive salvage therapy; however, treatment options remain limited and prognosis is poor.19 Depending on the patient’s cytogenetic profile, management can include HMA with or without VEN, glasdegib with LDAC, gilteritinib, ivosidenib or enasidenib, or gemtuzumab ozogamicin.9 Only a few studies have been published involving the r/rAML population not eligible for intensive salvage regimen, and guidelines are needed for this population.19 Thus, the ELN recommends that clinical trial enrollment be considered for patients with r/rAML.9

Management of Secondary AML or High-risk AML

Compared with de novo AML, both secondary AML (sAML) and therapy-related AML (tAML) have been associated with inferior outcomes. Factors that influence poor outcomes can include older age, comorbidities, persistent malignant disease or relapse of primary malignancy, treatment-induced depletion of hematopoietic reserves and/or prolonged myelosuppression, and genetic abnormalities, such as TP53 mutations.21

CPX-351 is a dual drug that contains cytarabine and daunorubicin.9,22 An open-label study (NCT01696084) compared CPX-351 with conventional cytarabine and daunorubicin (induction and consolidation therapy) in older patients (aged 60-75 years) with newly diagnosed high-risk/sAML who were considered fit for intensive therapy. The OS for CPX-351 was longer (9.56 vs 5.95 months) and the safety profiles were similar between the treatment groups.23 Patients achieving CR/CRi received up to 2 cycles of consolidation with CPX-351. An exploratory analysis of this subgroup revealed median OS was longer with CPX-351 consolidation (25.43 vs 8.53 months).22 Patients with TP53 mutations had poor treatment outcomes regardless of treatment arm, whereas patients with sAML-type mutations including spliceosome and chromatin modifier genes had longer OS with CPX-351 therapy.24 The 5-year results of this trial indicate that the survival benefit of CPX-351 was maintained.25 However, data from a retrospective review involving 136 patients with either sAML or AML with myelodysplasia-related changes revealed no difference in survival outcomes between patients treated with either HMA/VEN or CPX-351.26

Case Study: Elderly Woman With Newly Diagnosed AML

In 2018, Ms. W, age 69 years, was diagnosed with seropositive, non-erosive rheumatoid arthritis; she began methotrexate 17.5 mg per week split dosing in conjunction with oral folic acid 2 mg/d with varying doses based on symptoms. Her comorbidities included recurrent episodes of diverticulitis, hypertension, hypothyroidism, obstructive sleep apnea, and gastrointestinal reflux disease. On February 4, 2021, her methotrexate was increased to 20 mg and required intermittent prednisone tapers for flares. In November 2021, a blood test revealed she had a decreased white blood cell (WBC) count at 1.8 K/μL, and her methotrexate dose was decreased to 15 mg weekly. Despite the dose reduction, she had grade 3 neutropenia and anemia (WBC: 0.7 K/μL; HGB:10.5 g/dL) with a normal platelet count (PLT: 165,000/μL). Methotrexate was discontinued and leucovorin was initiated. She then had only modest improvement in her lab values and peripheral blood blasts. 

On March 17, 2022, she underwent a bone marrow biopsy and aspirate, which resulted in a diagnosis of AML. She had 55% blasts in a 90% cellular bone marrow with mild reticulin fibrosis and numerous circulating blasts. She was classified as having AML without maturation (FAB AML-M1). Flow cytometry detected a phenotypically abnormal population with CD45 expression and side scatter/forward scatter features of small-to-medium sized blasts, accounting for 23% of total cells. The chromosome analysis demonstrated a normal female karyotype in all 19 available metaphases. Polymerase chain reaction analysis was negative for FLT3-ITD, FLT3-TKD, and NPM1 mutations and positive for an IDH1 R132C missense mutation. The myeloid gene panel identified only a single pathogenic variant, IDH1 R132C (variant allele frequency [VAF] 21.2%), and a variant of unknown significance DNMT3A A575P (VAF 25.7%).

Noting that she does not have favorable risk features, we discussed treatment options. Although she is a candidate for curative therapy, the patient was not interested in pursuing allo-HCT. Her history of diverticulitis is concerning for tolerating intensive chemotherapy. In addition, her immunosuppressive therapy increases her risk for opportunistic infections. Based on the available data from the AGILE and VIALE studies and associated potential adverse reactions, she opted for starting treatment with AZA and IVO.

On March 31, 2022, she began receiving AZA 75 mg/m2 intravenous (IV) once daily days 1-7 and oral IVO 500 mg once daily continuously. She has received 12 cycles and has not needed transfusion. She has not had febrile neutropenia or symptoms of differentiation syndrome. On March 24, 2023, she underwent laparoscopic cholecystectomy, because an ultrasound revealed cholelithiasis, abnormal gallbladder wall thickening, and pericholecystic fluid. She was discharged home the following day and is continuing with AZA/ivosidenib.

Click to read more from 2023 Rare Diseases Report: Cancers

Harry Erba, MD, PhD
Defining “Unfit” for Intensive Chemotherapy

Within the last 40 years, younger fit patients have benefited from intensive chemotherapy regimens for acute myeloid leukemia (AML) with improved survival, and the possibility of long-term disease-free survival (DFS) (“cure”).1 Older patients are often considered too unfit for standard curative treatment with intensive induction chemotherapy followed by consolidation chemotherapy, allogeneic hematopoietic cell transplantation (allo-HCT), or both.2-4 Higher induction mortality and poor overall survival (OS) are associated with worse performance status, organ impairment, significant comorbidities, and declining cognitive function, all of which are more common with advancing age. Although the suggested criteria for determining unfitness have not been validated (Table 1), they can provide guidance in clinical practice.2-5

Table 1. Criteria to Define Unfitness for Intensive Chemotherapy to Treat AML

The National Comprehensive Cancer Network (NCCN) panel recommends the consideration of a patient’s performance status and comorbid conditions in addition to their age to determine a patient’s fitness for intensive induction therapy.6 Adverse disease features should also be considered, because disease biology may make intensive chemotherapy futile or inappropriate. For example, the mutational driver tumor protein p53 (TP53) appears at a higher frequency in older adults than younger adults and is associated with dismal outcomes even with intensive chemotherapy. Likewise, the spliceosome and chromatin modifier gene mutations are more common in older patients with AML and confer a worse OS with intensive therapy.6,7 Older unfit patients faced a difficult decision: proceed with intensive therapy with some possibility of long-term survival but risk of early mortality and significant toxicity, or opt for supportive care and palliative chemotherapy, such as the hypomethylating agents (HMAs) or low-dose cytarabine, with much shorter survival.

Guidelines for Treating Older Unfit Patients

Evidence-based guidelines for managing older adults with newly diagnosed AML were developed by the American Society of Hematology in 2020; however, these guidelines were released prior to the results of several clinical trials involving older patients with AML (Table 2).In 2022, the European LeukemiaNet (ELN) recommendations were updated to include new therapeutic agents that target specific mutations in genes such as tyrosine kinase 3 (FLT3), isocitrate dehydrogenase 1 (IDH1), isocitrate dehydrogenase 2 (IDH2), and B-cell lymphoma 2 (BCL2). Given the important effects of genetic aberrations on disease phenotype, treatment options, and outcomes, screening for genetic aberrations at diagnosis is now essential.9

Table 2. Treatment Landscape for Older Unfit Patients

The potential for clonal evolution leading to new actionable targets that were not present at diagnosis highlights the importance of reevaluation of genetic aberrations throughout clinical progression. Actionable targets can include mutations in IDH1/IDH2, FLT3-internal tandem duplication or FLT3 tyrosine kinase domain.9

Treatment Landscape

Since 2018, several therapeutic agents have been added to the treatment armamentarium that can induce longer-term complete remission (CR) for older unfit patients with newly diagnosed AML (Table 2).

Management of Primary AML With Less Intensive Induction Therapy

VIALE-A established a new standard of care for older unfit patients by demonstrating the benefit of adding the BCL2 inhibitor venetoclax (VEN) to azacitidine (AZA).2 VIALE-A demonstrated that the rate of CR plus CR with partial hematologic recovery (CRi) was 65% for VEN plus AZA and 18% for AZA. Most remissions in the AZA/VEN arm occurred rapidly in the first 2 cycles. The median survival improved from 9.6 months with AZA to 14.7 months with AZA/VEN. An improvement in survival with VEN and low-dose cytarabine also emerged in a 6-month post hoc analysis of the VIALE-C trial.10 Various other trials examining targeted therapies on specific mutations have provided mixed results in the front-line setting.13,14,18 It is important to note that a recent systematic review found that 12% to 25% of patients who were unfit for intensive therapy were successfully bridged to HCT.19

Management of Postremission Response

Patients with a longer duration of first remission have demonstrated better survival outcomes.15 Two trials have examined postremission therapy in the setting of prior intensive therapy. HOVON97 enrolled older patients who achieved CR/CRi after 2 cycles of intensive therapy to receive either AZA postremission or no further treatment. The proportion of patients with DFS at 12 months was greater in the AZA maintenance group than in the observation group (64% vs 42%), but significant DFS improvement did not translate into improved OS.20 QUAZAR AML-001 demonstrated that OS was longer for older patients receiving maintenance therapy with CC-486 (a non-bioequivalent oral formulation of AZA) vs placebo (24.7 vs 14.8 months).15 CC-486 was FDA-approved for maintenance therapy after intensive induction with or without consolidation in patients who are not candidates for allo-HCT. However, limited evidence exists specifically for postremission therapy in unfit patients who have received less intensive therapy. Continuation of the lower intensive therapy is recommended until disease progression.6 No data are available to support the use of oral AZA therapy alone for maintenance of remission following HMA/VEN-induced remissions.

Management of Relapsed and Refractory AML

Nearly 50% of patients with AML experience relapse and up to 40% may be refractory.19 Importantly, patients who were considered fit for intensive therapy may not remain so with relapsed or refractory AML (r/rAML), so patients should be evaluated for fitness for an intensive salvage regimen. Similar to assessing fitness for induction therapy, no standard definition of fitness exists for r/rAML.19

Disease control is the goal for patients with r/rAML who are unfit for intensive salvage therapy; however, treatment options remain limited and prognosis is poor.19 Depending on the patient’s cytogenetic profile, management can include HMA with or without VEN, glasdegib with LDAC, gilteritinib, ivosidenib or enasidenib, or gemtuzumab ozogamicin.9 Only a few studies have been published involving the r/rAML population not eligible for intensive salvage regimen, and guidelines are needed for this population.19 Thus, the ELN recommends that clinical trial enrollment be considered for patients with r/rAML.9

Management of Secondary AML or High-risk AML

Compared with de novo AML, both secondary AML (sAML) and therapy-related AML (tAML) have been associated with inferior outcomes. Factors that influence poor outcomes can include older age, comorbidities, persistent malignant disease or relapse of primary malignancy, treatment-induced depletion of hematopoietic reserves and/or prolonged myelosuppression, and genetic abnormalities, such as TP53 mutations.21

CPX-351 is a dual drug that contains cytarabine and daunorubicin.9,22 An open-label study (NCT01696084) compared CPX-351 with conventional cytarabine and daunorubicin (induction and consolidation therapy) in older patients (aged 60-75 years) with newly diagnosed high-risk/sAML who were considered fit for intensive therapy. The OS for CPX-351 was longer (9.56 vs 5.95 months) and the safety profiles were similar between the treatment groups.23 Patients achieving CR/CRi received up to 2 cycles of consolidation with CPX-351. An exploratory analysis of this subgroup revealed median OS was longer with CPX-351 consolidation (25.43 vs 8.53 months).22 Patients with TP53 mutations had poor treatment outcomes regardless of treatment arm, whereas patients with sAML-type mutations including spliceosome and chromatin modifier genes had longer OS with CPX-351 therapy.24 The 5-year results of this trial indicate that the survival benefit of CPX-351 was maintained.25 However, data from a retrospective review involving 136 patients with either sAML or AML with myelodysplasia-related changes revealed no difference in survival outcomes between patients treated with either HMA/VEN or CPX-351.26

Case Study: Elderly Woman With Newly Diagnosed AML

In 2018, Ms. W, age 69 years, was diagnosed with seropositive, non-erosive rheumatoid arthritis; she began methotrexate 17.5 mg per week split dosing in conjunction with oral folic acid 2 mg/d with varying doses based on symptoms. Her comorbidities included recurrent episodes of diverticulitis, hypertension, hypothyroidism, obstructive sleep apnea, and gastrointestinal reflux disease. On February 4, 2021, her methotrexate was increased to 20 mg and required intermittent prednisone tapers for flares. In November 2021, a blood test revealed she had a decreased white blood cell (WBC) count at 1.8 K/μL, and her methotrexate dose was decreased to 15 mg weekly. Despite the dose reduction, she had grade 3 neutropenia and anemia (WBC: 0.7 K/μL; HGB:10.5 g/dL) with a normal platelet count (PLT: 165,000/μL). Methotrexate was discontinued and leucovorin was initiated. She then had only modest improvement in her lab values and peripheral blood blasts. 

On March 17, 2022, she underwent a bone marrow biopsy and aspirate, which resulted in a diagnosis of AML. She had 55% blasts in a 90% cellular bone marrow with mild reticulin fibrosis and numerous circulating blasts. She was classified as having AML without maturation (FAB AML-M1). Flow cytometry detected a phenotypically abnormal population with CD45 expression and side scatter/forward scatter features of small-to-medium sized blasts, accounting for 23% of total cells. The chromosome analysis demonstrated a normal female karyotype in all 19 available metaphases. Polymerase chain reaction analysis was negative for FLT3-ITD, FLT3-TKD, and NPM1 mutations and positive for an IDH1 R132C missense mutation. The myeloid gene panel identified only a single pathogenic variant, IDH1 R132C (variant allele frequency [VAF] 21.2%), and a variant of unknown significance DNMT3A A575P (VAF 25.7%).

Noting that she does not have favorable risk features, we discussed treatment options. Although she is a candidate for curative therapy, the patient was not interested in pursuing allo-HCT. Her history of diverticulitis is concerning for tolerating intensive chemotherapy. In addition, her immunosuppressive therapy increases her risk for opportunistic infections. Based on the available data from the AGILE and VIALE studies and associated potential adverse reactions, she opted for starting treatment with AZA and IVO.

On March 31, 2022, she began receiving AZA 75 mg/m2 intravenous (IV) once daily days 1-7 and oral IVO 500 mg once daily continuously. She has received 12 cycles and has not needed transfusion. She has not had febrile neutropenia or symptoms of differentiation syndrome. On March 24, 2023, she underwent laparoscopic cholecystectomy, because an ultrasound revealed cholelithiasis, abnormal gallbladder wall thickening, and pericholecystic fluid. She was discharged home the following day and is continuing with AZA/ivosidenib.

Click to read more from 2023 Rare Diseases Report: Cancers

References
  1. Schlenk RF. Acute myeloid leukemia: introduction to a series highlighting progress and ongoing challenges. Haematologica. 2023;108(2):306-307. doi:10.3324/haematol.2022.280803
  2. DiNardo CD, Jonas BA, Pullarkat V, et al. Azacitidine and venetoclax in previously untreated acute myeloid leukemia. N Engl J Med. 2020;383(7):617-629. doi:10.1056/NEJMoa2012971
  3. DiNardo CD, Wei AH. How I treat acute myeloid leukemia in the era of new drugs. Blood. 2020;135(2):85-96. doi:10.1182/blood.2019001239
  4. Huerga-Domínguez S, Villar S, Prósper F, Alfonso-Piérola A. Updates on the management of acute myeloid leukemia. Cancers (Basel). 2022;14(19):4756. doi:10.3390/cancers14194756
  5. Ferrara F, Barosi G, Venditti A, et al. Consensus-based definition of unfitness to intensive and non-intensive chemotherapy in acute myeloid leukemia: a project of SIE, SIES and GITMO group on a new tool for therapy decision making. Leukemia. 2013;27(5):997-999. doi:10.1038/leu.2012.303
  6. Tallman MS, Wang ES, Altman JK, et al. Acute myeloid leukemia, version 3.2019, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2019;17(6):721-749. doi:10.6004/jnccn.2019.0028
  7. Burd A, Levine RL, Ruppert AS, et al. Precision medicine treatment in acute myeloid leukemia using prospective genomic profiling: feasibility and preliminary efficacy of the Beat AML Master Trial. Nat Med. 2020;26(12):1852-1858. doi:10.1038/s41591-020-1089-8
  8. Sekeres MA, Guyatt G, Abel G, et al. American Society of Hematology 2020 guidelines for treating newly diagnosed acute myeloid leukemia in older adults. Blood Adv. 2020;4(15):3528-3549. doi:10.1182/bloodadvances.2020001920
  9. Döhner H, Wei AH, Appelbaum FR, et al. Diagnosis and management of AML in adults: 2022 recommendations from an international expert panel on behalf of the ELN. Blood. 2022;140(12):1345-1377. doi:10.1182/blood.2022016867
  10. Wei AH, Montesinos P, Ivanov V, et al. Venetoclax plus LDAC for newly diagnosed AML ineligible for intensive chemotherapy: a phase 3 randomized placebo-controlled trial. Blood. 2020;135(24):2137-2145. doi:10.1182/blood.2020004856
  11. Amadori S, Suciu S, Selleslag D, et al. Gemtuzumab ozogamicin versus best supportive care in older patients with newly diagnosed acute myeloid leukemia unsuitable for intensive chemotherapy: results of the randomized phase III EORTC-GIMEMA AML-19 trial. J Clin Oncol. 2016;34(9):972-979. doi:10.1200/JCO.2015.64.0060
  12. Cortes JE, Heidel FH, Hellmann A, et al. Randomized comparison of low dose cytarabine with or without glasdegib in patients with newly diagnosed acute myeloid leukemia or high-risk myelodysplastic syndrome. Leukemia. 2019;33(2):379-389. doi:10.1038/s41375-018-0312-9
  13. Montesinos P, Recher C, Vives S, et al. Ivosidenib and azacitidine in IDH1-mutated acute myeloid leukemia. N Engl J Med. 2022;386(16):1519-1531. doi:10.1056/NEJMoa2117344
  14. Wang ES, Montesinos P, Minden MD, et al. Phase 3 trial of gilteritinib plus azacitidine vs azacitidine for newly diagnosed FLT3mut+ AML ineligible for intensive chemotherapy. Blood. 2022;140(17):1845-1857. doi:10.1182/blood.2021014586
  15. Wei AH, Döhner H, Pocock C, et al; QUAZAR AML-001 Trial Investigators. Oral azacitidine maintenance therapy for acute myeloid leukemia in first remission. N Engl J Med. 2020;383(26):2526-2537. doi:10.1056/NEJMoa2004444
  16. Perl AE, Martinelli G, Cortes JE, et al. Gilteritinib or chemotherapy for relapsed or refractory FLT3-mutated AML. N Engl J Med. 2019;381(18):1728-1740. doi:10.1056/NEJMoa1902688
  17. Konopleva MY, Röllig C, Cavenagh J, et al. Idasanutlin plus cytarabine in relapsed or refractory acute myeloid leukemia: results of the MIRROS trial. Blood Adv. 2022;6(14):4147-4156. doi:10.1182/bloodadvances.2021006303
  18. Pollyea DA, DiNardo CD, Arellano ML, et al. Impact of venetoclax and azacitidine in treatment-naïve patients with acute myeloid leukemia and IDH1/2 mutations. Clin Cancer Res. 2022;28(13):2753-2761. doi:10.1158/1078-0432.CCR-21-3467
  19. Russell-Smith TA, Gurskyte L, Muresan B, et al. Efficacy of non-intensive therapies approved for relapsed/refractory acute myeloid leukemia: a systematic literature review. Future Oncol. 2022;18(16):2029-2039. doi:10.2217/fon-2021-1355
  20. Huls G, Chitu DA, Havelange V, et al; Dutch-Belgian Hemato-Oncology Cooperative Group (HOVON). Azacitidine maintenance after intensive chemotherapy improves DFS in older AML patients. Blood. 2019;133(13):1457-1464. doi:10.1182/blood-2018-10-879866
  21. Granfeldt Østgård LS, Medeiros BC, Sengeløv H, et al. Epidemiology and clinical significance of secondary and therapy-related acute myeloid leukemia: a national population-based cohort study. J Clin Oncol. 2015;33(31):3641-3649. doi:10.1200/JCO.2014.60.0890
  22. Kolitz JE, Strickland SA, Cortes JE, et al. Consolidation outcomes in CPX-351 versus cytarabine/daunorubicin-treated older patients with high-risk/secondary acute myeloid leukemia. Leuk Lymphoma. 2020;61(3):631-640. doi:10.1080/1042819.2019.1688320
  23. Lancet JE, Uy GL, Cortes JE, et al. CPX-351 (cytarabine and daunorubicin) liposome for injection versus conventional cytarabine plus daunorubicin in older patients with newly diagnosed secondary acute myeloid leukemia. J Clin Oncol. 2018;36(26):2684-2692. doi:10.1200/JCO.2017.77.6112
  24. Lindsley RC, Gibson CJ, Murdock HM, et al. Genetic characteristics and outcomes by mutation status in a phase 3 study of CPX-351 versus 7+3 in older adults with newly diagnosed, high-risk/secondary acute myeloid leukemia (AML). Blood. 2019;134(suppl 1):15. doi:10.1182/blood-2019-124500
  25. Lancet JE, Uy GL, Newell LF, et al. CPX-351 versus 7+3 cytarabine and daunorubicin chemotherapy in older adults with newly diagnosed high-risk or secondary acute myeloid leukaemia: 5-year results of a randomised, open-label, multicentre, phase 3 trial. Lancet Haematol. 2021;8(7):e481-e491. doi:10.1016/S2352-3026(21)00134-4
  26. Alharthy H, Alkaabba F, Williams M, et al. Outcomes of newly diagnosed therapy-related AML and AML with myelodysplasia-related changes treated with 7+3, hypomethylating agents with or without venetoclax and CPX-351: a retrospective cohort study. Blood. 2022;140(suppl 1):9025-9026. doi:10.1182/blood-2022-170688
References
  1. Schlenk RF. Acute myeloid leukemia: introduction to a series highlighting progress and ongoing challenges. Haematologica. 2023;108(2):306-307. doi:10.3324/haematol.2022.280803
  2. DiNardo CD, Jonas BA, Pullarkat V, et al. Azacitidine and venetoclax in previously untreated acute myeloid leukemia. N Engl J Med. 2020;383(7):617-629. doi:10.1056/NEJMoa2012971
  3. DiNardo CD, Wei AH. How I treat acute myeloid leukemia in the era of new drugs. Blood. 2020;135(2):85-96. doi:10.1182/blood.2019001239
  4. Huerga-Domínguez S, Villar S, Prósper F, Alfonso-Piérola A. Updates on the management of acute myeloid leukemia. Cancers (Basel). 2022;14(19):4756. doi:10.3390/cancers14194756
  5. Ferrara F, Barosi G, Venditti A, et al. Consensus-based definition of unfitness to intensive and non-intensive chemotherapy in acute myeloid leukemia: a project of SIE, SIES and GITMO group on a new tool for therapy decision making. Leukemia. 2013;27(5):997-999. doi:10.1038/leu.2012.303
  6. Tallman MS, Wang ES, Altman JK, et al. Acute myeloid leukemia, version 3.2019, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2019;17(6):721-749. doi:10.6004/jnccn.2019.0028
  7. Burd A, Levine RL, Ruppert AS, et al. Precision medicine treatment in acute myeloid leukemia using prospective genomic profiling: feasibility and preliminary efficacy of the Beat AML Master Trial. Nat Med. 2020;26(12):1852-1858. doi:10.1038/s41591-020-1089-8
  8. Sekeres MA, Guyatt G, Abel G, et al. American Society of Hematology 2020 guidelines for treating newly diagnosed acute myeloid leukemia in older adults. Blood Adv. 2020;4(15):3528-3549. doi:10.1182/bloodadvances.2020001920
  9. Döhner H, Wei AH, Appelbaum FR, et al. Diagnosis and management of AML in adults: 2022 recommendations from an international expert panel on behalf of the ELN. Blood. 2022;140(12):1345-1377. doi:10.1182/blood.2022016867
  10. Wei AH, Montesinos P, Ivanov V, et al. Venetoclax plus LDAC for newly diagnosed AML ineligible for intensive chemotherapy: a phase 3 randomized placebo-controlled trial. Blood. 2020;135(24):2137-2145. doi:10.1182/blood.2020004856
  11. Amadori S, Suciu S, Selleslag D, et al. Gemtuzumab ozogamicin versus best supportive care in older patients with newly diagnosed acute myeloid leukemia unsuitable for intensive chemotherapy: results of the randomized phase III EORTC-GIMEMA AML-19 trial. J Clin Oncol. 2016;34(9):972-979. doi:10.1200/JCO.2015.64.0060
  12. Cortes JE, Heidel FH, Hellmann A, et al. Randomized comparison of low dose cytarabine with or without glasdegib in patients with newly diagnosed acute myeloid leukemia or high-risk myelodysplastic syndrome. Leukemia. 2019;33(2):379-389. doi:10.1038/s41375-018-0312-9
  13. Montesinos P, Recher C, Vives S, et al. Ivosidenib and azacitidine in IDH1-mutated acute myeloid leukemia. N Engl J Med. 2022;386(16):1519-1531. doi:10.1056/NEJMoa2117344
  14. Wang ES, Montesinos P, Minden MD, et al. Phase 3 trial of gilteritinib plus azacitidine vs azacitidine for newly diagnosed FLT3mut+ AML ineligible for intensive chemotherapy. Blood. 2022;140(17):1845-1857. doi:10.1182/blood.2021014586
  15. Wei AH, Döhner H, Pocock C, et al; QUAZAR AML-001 Trial Investigators. Oral azacitidine maintenance therapy for acute myeloid leukemia in first remission. N Engl J Med. 2020;383(26):2526-2537. doi:10.1056/NEJMoa2004444
  16. Perl AE, Martinelli G, Cortes JE, et al. Gilteritinib or chemotherapy for relapsed or refractory FLT3-mutated AML. N Engl J Med. 2019;381(18):1728-1740. doi:10.1056/NEJMoa1902688
  17. Konopleva MY, Röllig C, Cavenagh J, et al. Idasanutlin plus cytarabine in relapsed or refractory acute myeloid leukemia: results of the MIRROS trial. Blood Adv. 2022;6(14):4147-4156. doi:10.1182/bloodadvances.2021006303
  18. Pollyea DA, DiNardo CD, Arellano ML, et al. Impact of venetoclax and azacitidine in treatment-naïve patients with acute myeloid leukemia and IDH1/2 mutations. Clin Cancer Res. 2022;28(13):2753-2761. doi:10.1158/1078-0432.CCR-21-3467
  19. Russell-Smith TA, Gurskyte L, Muresan B, et al. Efficacy of non-intensive therapies approved for relapsed/refractory acute myeloid leukemia: a systematic literature review. Future Oncol. 2022;18(16):2029-2039. doi:10.2217/fon-2021-1355
  20. Huls G, Chitu DA, Havelange V, et al; Dutch-Belgian Hemato-Oncology Cooperative Group (HOVON). Azacitidine maintenance after intensive chemotherapy improves DFS in older AML patients. Blood. 2019;133(13):1457-1464. doi:10.1182/blood-2018-10-879866
  21. Granfeldt Østgård LS, Medeiros BC, Sengeløv H, et al. Epidemiology and clinical significance of secondary and therapy-related acute myeloid leukemia: a national population-based cohort study. J Clin Oncol. 2015;33(31):3641-3649. doi:10.1200/JCO.2014.60.0890
  22. Kolitz JE, Strickland SA, Cortes JE, et al. Consolidation outcomes in CPX-351 versus cytarabine/daunorubicin-treated older patients with high-risk/secondary acute myeloid leukemia. Leuk Lymphoma. 2020;61(3):631-640. doi:10.1080/1042819.2019.1688320
  23. Lancet JE, Uy GL, Cortes JE, et al. CPX-351 (cytarabine and daunorubicin) liposome for injection versus conventional cytarabine plus daunorubicin in older patients with newly diagnosed secondary acute myeloid leukemia. J Clin Oncol. 2018;36(26):2684-2692. doi:10.1200/JCO.2017.77.6112
  24. Lindsley RC, Gibson CJ, Murdock HM, et al. Genetic characteristics and outcomes by mutation status in a phase 3 study of CPX-351 versus 7+3 in older adults with newly diagnosed, high-risk/secondary acute myeloid leukemia (AML). Blood. 2019;134(suppl 1):15. doi:10.1182/blood-2019-124500
  25. Lancet JE, Uy GL, Newell LF, et al. CPX-351 versus 7+3 cytarabine and daunorubicin chemotherapy in older adults with newly diagnosed high-risk or secondary acute myeloid leukaemia: 5-year results of a randomised, open-label, multicentre, phase 3 trial. Lancet Haematol. 2021;8(7):e481-e491. doi:10.1016/S2352-3026(21)00134-4
  26. Alharthy H, Alkaabba F, Williams M, et al. Outcomes of newly diagnosed therapy-related AML and AML with myelodysplasia-related changes treated with 7+3, hypomethylating agents with or without venetoclax and CPX-351: a retrospective cohort study. Blood. 2022;140(suppl 1):9025-9026. doi:10.1182/blood-2022-170688
Publications
Publications
Topics
Article Type
Display Headline
Treatment Needs of Older Adults With Newly Diagnosed Acute Myeloid Leukemia
Display Headline
Treatment Needs of Older Adults With Newly Diagnosed Acute Myeloid Leukemia
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Thu, 06/22/2023 - 22:00
Un-Gate On Date
Thu, 06/22/2023 - 22:00
Use ProPublica
CFC Schedule Remove Status
Thu, 06/22/2023 - 22:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Advances in Management of Relapsed/Refractory Hairy Cell Leukemia

Article Type
Changed
Mon, 06/26/2023 - 12:31
Display Headline
Advances in Management of Relapsed/Refractory Hairy Cell Leukemia

Robert J. Kreitman, MD
Hairy cell leukemia (HCL) is an indolent, low-grade B-cell lymphoid malignancy that typically presents with fatigue, pancytopenia, and splenomegaly.1,2 It is a rare disease, with an estimated 1,200 new cases of HCL diagnosed annually in the United States.1 Demographically, HCL is a disease of older adults (median age at diagnosis, 58 years), and is more commonly found in men than women and in White individuals compared with other ethnic or racial backgrounds.3,4 Environmental or occupational exposure to toxic substances, pesticides, ionizing radiation, and petroleum products may be linked to increased risk for HCL development.1,4


Pathophysiology

HCL develops from activated, mature memory B-cells that, in most cases, have the acquired mutation in BRAF V600E, which is present in 80% to 90% of patients with classic HCL.1,3,5 BRAF is an integral part of the RAS-BRAF-MEK-ERK cellular pathway that transmits growth factor signals from the cell surface to the nucleus to regulate cell growth and proliferation.Mutated BRAF V600E continuously activates BRAF kinase and downstream signaling, resulting in enhanced HCL cell survival and unchecked proliferation.3

Variant HCL (HCLv) is a separate, more virulent disease that lacks BRAF V600E mutation and CD25 expression on flow cytometry.1,7-9 Patients with HCLv have a worse prognosis and poor responses to front-line purine analogs, and a higher proportion of these patients carry the unmutated immunoglobulin heavy chain variable (IGHV) gene (54% vs 17% in HCL).1,10,11 About 30% to 50% have wild-type BRAF and activating mutations in MAP2K1, which encodes aberrant MEK downstream of BRAF.10,12

Most patients with HCL have somatic mutations in the IGHV gene.3,13,14 Patients with unmutated IGHV4-34 and wildtype BRAF have an aggressive form of the disease, even if the HCL cells express CD25 as in classic HCL.1,15 HCL in patients with unmutated IGHV is often refractory to purine analogs and these patients have poor prognosis and rapid progression.16 Other identified mutations include CDKN1B in HCL and MAP2K1 and CCNC3 in HCLv.2

Signs and Symptoms

In many cases, HCL is asymptomatic, and diagnosed when pancytopenia, monocytopenia, and leukopenia are discovered on unrelated blood work.2,3,11 Monocytopenia is a specific presentation of HCL, but not HCLv.11 Typical systemic symptoms include unexplained weight loss and extreme fatigue (80%).1,3 Other symptoms can include fever, recurrent infections, night sweats, splenomegaly and related pain or abdominal fullness, hepatomegaly, and bleeding or bruising due to thrombocytopenia.1,3 Splenomegaly is associated with advanced disease.11

Up to 30% of patients may present with autoimmune disorders such as vasculitis or psoriasis. Although skin involvement is rare with HCL, 10% to 12% of patients will have dermatologic symptoms either due to recurrent infection or autoimmune reactions.1,2 Skin reactions include localized or generalized maculopapular rash, pyoderma gangrenosum (which may be severe), and recurrent bacterial or viral skin infections.17

Diagnosis

After complete history and physical examination, a diagnosis of HCL is usually made based on flow cytometry for immunophenotyping and molecular testing for BRAF V600E (Table 1).2,17

Table 1. Essential Tests for HCL Diagnosis

Disease-related fibrosis may impede bone marrow aspiration, and trephine biopsy should be done to make the diagnosis.11 On morphologic examination, HCL cells are small- to medium-sized, with round, oval, or indented, well-defined nuclei. Cytoplasm is pale blue, and cells have small cytoplasmic projections (Figure 1).2,18

©Getty Images/Kateryna Kon/Science Photo Library
Figure 1. Typical Appearance of Hairy Cell Leukemia

On flow cytometry, HCL is positive for B-cell antigens (CD19, CD20, CD22), as well as antigens specific to the disease (CD11c, CD25, CD103, CD123), and by immunohistochemistry (IHC) for cyclin D1 and annexin-A1. CD20, CD123, and CD200 are bright in HCL. The presence of T-cell marker CD103 on B-cells indicates HCL.1-3 HCLv, in contrast, is positive for CD11c and CD103, but usually negative for CD25, CD123, and annexin-A1.2,19

BRAF V600E mutation can be identified using droplet digital polymerase chain reaction (PCR), next-generation molecular sequencing, or IHC with a VE1 stain.3,11 IHC for CD20, annexin-1, and VE1 establish the diagnosis, but also are useful in determining the extent to which leukemic cells have infiltrated bone marrow.11

Differential diagnosis of HCL includes HCL variants, splenic marginal zone lymphoma, and splenic diffuse red pulp small B-cell lymphoma.7,11

Indications for Treatment and Criteria for Response

Over time, about 90% of patients with HCL will require treatment. However, not all such patients will require urgent or immediate treatment, and some can be managed with observation and close monitoring.1,11 The indications for initiating treatment generally are systemic symptoms and significant pancytopenia (Table 2).2,11

Table 3. Indications for Treatment of HCL

The optimal response with treatment of HCL is complete response (CR) without minimal residual disease (MRD-free), which minimizes the risk for relapse.1,11 Hematologic and molecular response is assessed using peripheral blood samples; physical examination, ultrasound, computed tomography, or magnetic resonance imaging is used to determine response in lymph nodes, spleen, or liver.1 MRD-free is defined by the absence of HCL cells by the chosen method (IHC, flow cytometry, or PCR).20 Bone marrow aspirate flow cytometry is the most sensitive standard test for MRD detection.1Table 3 summarizes response criteria for HCL.2,11

Table 3. Indications for Treatment of HCL

Initial Treatment of HCL

The purine nucleoside analogs (PNAs) cladribine (± rituximab) and pentostatin are widely recommended for initial treatment.1,2,11 As monotherapy, cladribine and pentostatin are considered similarly effective, with CR in 70% to 90% of patients and durations of response > 10 years.1 Adding the anti-CD20 monoclonal antibody rituximab in 8 weekly doses starting the first day of front-line cladribine (CDAR) improves remission, MRD-free rates, and duration of response (94% MRD-free at 96 months), with minimal added toxicity.21 Rituximab is often added 4 weeks after cladribine, which offers more convenience, an equally high CR rate of 100%, and a 76% MRD-free rate at 3 months.11 Bone marrow biopsy should be delayed for 4 to 6 months to allow a full response to develop with cladribine.1,11

Daily (intravenous or subcutaneous) and weekly cladribine are equally safe and effective.2,11 Pentostatin is administered intravenously every 2 weeks for 3 to 6 months, allowing time for hematologic recovery between doses.1,11 Patient factors to consider when choosing treatment include baseline neutropenia, patient preference, and comorbidities.

Toxicities of PNAs include neutropenia and fever, which typically occur during the first month of treatment and are more frequent in patients with baseline severe neutropenia; T-cell recovery may take years.1 CDAR is associated with higher transient thrombocytopenia, but faster platelet and neutrophil recovery at 4 weeks than cladribine alone.21 Both therapies are immunosuppressive. Patients should be evaluated for existing infections and watched for new infections during treatment. Control of active infection prior to treatment initiation is required.11,23

Patients with confirmed BRAF V600E mutation are candidates for vemurafenib if they are unable to tolerate a PNA, have an active infection, or would like effective vaccinations.2,23-25

Treatment at Relapse

At suspected HCL relapse, patients should be evaluated to determine whether cytopenia is due to recurrent disease or lingering effects from prior treatment. Use of successive flow cytometry over time can clarify whether symptoms are related to disease and need interventional treatment, or will resolve with additional time.1

Patients who have an HCL relapse after initial therapy with cladribine or pentostatin may be candidates for re-treatment with the same or alternate PNA plus rituximab.2 Rituximab
monotherapy has been used for patients unable to tolerate PNA but yields CR rates as low as 13%.26 Repeated courses of PNA therapy yield lower rates and durations of response with each course.1,2

For patients with primary refractory disease (less than CR with initial therapy) or relapse within 2 years of initial therapy, treatment with the BRAF V600E inhibitor vemurafenib off-label, with or without rituximab, is an option.2,5 In HCL, vemurafenib for patients with relapsed or refractory disease achieved CR in 35% and 42% in 2 small trials (N = 54). Relapse-free survival among people with CR was 19 months in 1 of the trials.27 Vemurafenib plus rituximab achieved CR in 87% of patients with relapsed or refractory HCL, and an MRD-free CR rate of 57%. Among patients with CR, 85% were relapse-free at a median follow-up of 34 months.5 Treatment with vemurafenib is not myelotoxic—an advantage for HCL patients. Adverse effects with vemurafenib are often manageable with dose reductions, if needed. A specific concern with vemurafenib is the potential development of secondary skin cancers.5,27,28

Novel Targeted Options and Recommended Use

Promising alternatives for patients with relapsed or refractory HCL include combined BRAF and MEK inhibitors and the Bruton tyrosine kinase (BTK) inhibitor ibrutinib. The concept of BRAF/MEK inhibition was validated in studies with BRAF-mutated melanoma, in which dabrafenib plus trametinib (the MEK inhibitor) improved overall survival (OS) with less toxicity and better quality of life than vemurafenib.1,29 In a phase 2 trial in HCL, dabrafenib monotherapy demonstrated an overall response rate (ORR) of 80%, including 30% CR.30 In a subsequent phase 2 trial, dabrafenib combined with trametinib was evaluated in refractory or late relapsed HCL. Among 55 enrolled patients, objective response rate was 89%, including 65.5% CR. Nine of 36 patients with CR were MRD-free. Among responding patients, duration of response was 97.7% at 24 months.31 The most common grade ≥ 3 toxicities were hyperglycemia, pyrexia, neutropenia, and pneumonia. Secondary skin cancers were seen in about 5% of patients.31

BRAF/MEK inhibitor combinations in HCL offer effective therapy with less myelosuppression than PNAs, making them useful for patients with or at risk for infection.23 Their use in HCL is off-label, as they currently are approved for treatment of BRAF-mutated melanoma and some other tumors.32 A study of encorafenib (a BRAF inhibitor) combined with binimetinib (a MEK inhibitor) is ongoing (Table 4).32

Table 4. Currently Recruiting Clinical Trials Specifically for HCL

Ibrutinib interrupts B-cell receptor signaling to stop tumor cell growth. In a phase 2 trial, patients with relapsed or refractory HCL or HCLv were treated with once-daily oral ibrutinib. Best ORR was 54% (19% CR; 3% MRD-free). Despite the low CR rate, 3-year progression-free survival with ibrutinib was 73% and OS was 85%. Treatment was well-tolerated; cytopenia (including 22% grade ≥ 3 thrombocytopenia and neutropenia) and diarrhea were frequent toxicities.33

Moxetumomab pasudotox is a novel CD22-targeted antibody fused with protein toxin that interrupts protein synthesis in tumor cells.1 As treatment, it was studied in a phase 3 trial of relapsed HCL in heavily pretreated patients, and achieved a CR rate of 41%, including 36% durable CR.34 Although FDA-approved for relapsed or refractory HCL, the drug is being discontinued due to business decisions, not safety or efficacy concerns.2 It is notable that many types of B-cell lymphoma also express CD22.35

Enrollment in a clinical trial to study possible treatment advances is recommended by the National Comprehensive Cancer Network (NCCN) at first and subsequent relapses of HCL for appropriate patients.Figure 2 summarizes an approach to treatment choice and sequencing for patients with HCL.

Figure 2. Standard & Investigational Treatment of HCL/HCLv

Supportive Care

Patients being treated for HCL should have supportive care to manage adverse effects of their disease. Such care includes prophylaxis against herpes virus if CD4+ T cells < 200 cells/μL and other prophylactic vaccinations to hepatitis B virus, COVID-19 and Influenza. Patients with neutropeni may require broad-spectrum antibacterial prophylaxis or neutrophil growth factors if neutropenic fever develops. Blood product support is recommended if needed.2 Assessment of anti-COVID-19 antibodies is recommended to optimize immunity, particularly prior to beginning anti-CD20 antibody therapy like rituximab.23

Unmet Needs

Despite improvements in response and survival with newer therapies, not all patients with HCL benefit from these advances. Unmet needs are finding optimal treatment for patients with HCLv, despite some success with MEK inhibitors, and for patients with BRAF mutations other than V600E, who have few options beyond PNAs and rituximab.

Click to read more from 2023 Rare Diseases Report: Cancers

References
  1. Kreitman RJ, Arons E. Diagnosis and treatment of hairy cell leukemia as the COVID-19 pandemic continues. Blood Rev. 2022;51:100888. doi:10.1016/j.blre.2021.100888
  2. National Comprehensive Cancer Network. NCCN clinical practice guideline in oncology: hairy cell leukemia. Version 1.2023. Published August 30, 2022. Accessed March 16, 2023. https://www.nccn.org/professionals/physician_gls/pdf/hairy_cell.pdf
  3. Janus A, Robak T. Hairy cell leukemia. In: Li W, ed. Leukemia [Internet]. Brisbane: Exon Publications; 2022:chap3. Accessed February 16, 2023. doi:10.36255/exon-publications-leukemia-hairy-cell-leukemia
  4. Tadmor T, Polliack A. Epidemiology and environmental risk in hairy cell leukemia. Best Pract Res Clin Haematol. 2015;28(4):175-179. doi:10.1016/j.beha.2015.10.014
  5. Tiacci E, De Carolis L, Simonetti E, et al. Vemurafenib plus rituximab in refractory or relapsed hairy-cell leukemia. N Engl J Med. 2021;384(19):1810-1823. doi:10.1056/NEJMoa20312986
  6. Falini B, Martelli MP, Tiacci E. BRAF V600E mutation in hairy cell leukemia: from bench to bedside. Blood. 2016;128(15):1918-1927. doi:10.1182/blood-2016-07-418434
  7. Matutes E. Diagnostic and therapeutic challenges in hairy cell leukemia-variant: where are we in 2021? Expert Rev Hematol. 2021;14(4):355-363. doi:10.1080/17474086.2021.1908121
  8. Cawley JC, Burns GF, Hayhoe FG. A chronic lymphoproliferative disorder with distinctive features: a distinct variant of hairy-cell leukaemia. Leuk Res. 1980;4(6):547-559. doi:10.1016/0145-2126(80)90066-1
  9. Xi L, Arons E, Navarro W, et al. Both variant and IGHV4-34-expressing hairy cell leukemia lack the BRAF V600E mutation. Blood. 2012;119(14):3330-3332. doi:10.1182/blood-2011-09-379339
  10. Durham BH, Getta B, Dietrich S, et al. Genomic analysis of hairy cell leukemia identifies novel recurrent genetic alterations. Blood. 2017;130(14):1644-1648. doi:10.1182/blood-2017-01-76510711
  11. Grever MR, Abdel-Wahab O, Andritsos LA, et al. Consensus guidelines for the diagnosis and management of patients with hairy cell leukemia. Blood. 2017;129(5):553-560. doi:10.1182/blood-2016-01-689422
  12. Waterfall JJ, Arons E, Walker RL, et al. High prevalence of MAP2K1 mutations in variant and IGHV4-34-expressing hairy-cell leukemias. Nat Genet. 2014;46(1):8-10. doi:10.1038/ng.2828
  13. Arons E, Sunshine J, Suntum T, Kreitman RJ. Somatic hypermutation and VH gene usage in hairy cell leukaemia. Br J Haematol. 2006;133(5):504-512. doi:10.1111/j.1365-2141.2006.06066.x
  14. Arons E, Roth L, Sapolsky J, Suntum T, Stetler-Stevenson M, Kreitman RJ. Evidence of canonical somatic hypermutation in hairy cell leukemia. Blood. 2011;117(18):4844-4851. doi:10.1182/blood-2010-11-316737
  15. Arons E, Suntum T, Stetler-Stevenson M, Kreitman RJ. VH4-34+ hairy cell leukemia, a new variant with poor prognosis despite standard therapy. Blood. 2009;114(21):4687-4695. doi:10.1182/blood-2009-01-201731
  16. Forconi F, Sozzi E, Cencini E, et al. Hairy cell leukemias with unmutated IGHV genes define the minor subset refractory to single-agent cladribine and with more aggressive behavior. Blood. 2009;114(21):4696-4702. doi:10.1182/blood-2009-03-212449
  17. Robak E, Jesionek-Kupnicka D, Robak T. Skin changes in hairy cell leukemia. Ann Hematol. 2021;100(3):615-625. doi:10.1007/s00277-020-04349-z
  18. Bouroncle BA. Thirty-five years in the progress of hairy cell leukemia. Leuk Lymphoma. 1994;14(suppl 1):1-12. https://pubmed.ncbi.nlm.nih.gov/7820038/
  19. Falini B, Tiacci E, Liso A, et al. Simple diagnostic assay for hairy cell leukaemia by immunocytochemical detection of annexin A1 (ANXA1). Lancet. 2004;363(9424): 1869-1870. doi:10.1016/S0140-6736(04)16356-3
  20. Robak T, Robak P. Measurable residual disease in hairy cell leukemia: technical considerations and clinical significance. Front Oncol. 2022;12:976374. doi:10.3389/fonc.2022.976374
  21. Chihara D, Arons E, Stetler-Stevenson M, et al. Randomized phase II study of first-line cladribine with concurrent or delayed rituximab in patients with hairy cell leukemia. J Clin Oncol. 2020;38(14):1527-1538. doi:10.1200/JCO.19.02250
  22. Chihara D, Kantarjian H, O’Brien S, et al. Long-term durable remission by cladribine followed by rituximab in patients with hairy cell leukaemia: update of a phase II trial. Br J Haematol. 2016;174(5):760-766. doi:10.1111/bjh.14129
  23. Grever M, Andritsos L, Banerji V, et al. Hairy cell leukemia and COVID-19 adaptation of treatment guidelines. Leukemia. 2021;35(7):1864-1872. doi:10.1038/s41375-021-01257-7
  24. Konrat J, Rösler W, Roiss M, et al. BRAF inhibitor treatment of classical hairy cell leukemia allows successful vaccination against SARS-CoV-2. Ann Hematol. 2023;102(2):403-406. doi:10.1007/s00277-022-05026-z
  25. Park JH, Shukla M, Salcedo JM, et al. First-line chemo-free therapy with the BRAF inhibitor vemurafenib combined with obinutuzumab is effective in patients with HCL. Blood. 2019;134(suppl 1):Abstract 3998. https://doi.org/10.1182/blood-2019-124478
  26. Nieva J, Bethel K, Saven A. Phase 2 study of rituximab in the treatment of cladribine-failed patients with hairy cell leukemia. Blood. 2003;102(3):810-813. doi:10.1182/blood-2003-01-0014
  27. Tiacci E, Park JH, De Carolis L, et al. Targeting mutant BRAF in relapsed or refractory hairy-cell leukemia. N Engl J Med. 2015;373(18):1733-1747. doi:10.1056/NEJMoa1506583
  28. Maitre E, Paillassa J, Troussard X. Novel targeted treatments in hairy cell leukemia and other hairy cell-like disorders. Front Oncol. 2022;12:1068981. doi:10.3389/fonc.2022.1068981
  29. Grob JJ, Amonkar MM, Karaszewska B, et al. Comparison of dabrafenib and trametinib combination therapy with vemurafenib monotherapy on health-related quality of life in patients with unresectable or metastatic cutaneous BRAF Val600-mutation-positive melanoma (COMBI-v): results of a phase 3, open-label, randomised trial. Lancet Oncol. 2015;16(13):1389-1398. doi:10.1016/S1470-2045(15)00087-X
  30. Tiacci E, De Carolis L, Simonetti E, et al. Safety and efficacy of the BRAF inhibitor dabrafenib in relapsed or refractory hairy cell leukemia: a pilot phase-2 clinical trial. Leukemia. 2021;35(11):3314-3318. doi:10.1038/s41375-021-01210-8
  31. Kreitman RJ, Moreau P, Ravandi F, et al. Dabrafenib plus trametinib in patients with relapsed/refractory BRAF V600E mutation-positive hairy cell leukemia. Blood. 2023;141(9):996-1006. doi:10.1182/blood.2021013658
  32. Adashek JJ, Menta AK, Reddy NK, Desai AP, Roszik J, Subbiah V. Tissue agnostic activity of BRAF plus MEK inhibitor in BRAF V600E-mutated tumors. Mol Cancer Ther. 2022;21(6):871-878. doi:10.1158/1535-7163.MCT-21-0950
  33. Rogers KA, Andritsos LA, Wei L, et al. Phase 2 study of ibrutinib in classic and variant hairy cell leukemia. Blood. 2021;137(25):3473-3483. doi:10.1182/blood.2020009688
  34. Kreitman RJ, Dearden C, Zinzani PL, et al; Study 1053 investigators. Moxetumomab pasudotox in heavily pre-treated patients with relapsed/refractory hairy cell leukemia (HCL): long-term follow-up from the pivotal trial. J Hematol Oncol. 2021;14(1):35. doi:10.1186/s13045-020-01004-y
  35. Leonard JP, Goldenberg DM. Preclinical and clinical evaluation of epratuzumab (anti-CD22 IgG) in B-cell malignancies. Oncogene. 2007;26(25):3704-3713. doi:10.1038/sj.onc.1210370
Author and Disclosure Information

Robert J. Kreitman, MD
Senior Investigator
National Cancer Institute
National Institutes of Health
Bethesda, MD

Robert J. Kreitman, MD, has disclosed the following relevant financial relationships:

Coinventor on the NIH patent for Moxetumomab Pasudotox.

Receives research support and/or investigation drugs for trials from AstraZeneca, Pfizer, Novartis, Teva, and Genentech.

Dr. Robert J. Kreitman contributed to this article in his personal capacity. The views expressed are his own and do not necessarily represent tthe views of the National Institutes of Health or the United States Government.

Publications
Topics
Author and Disclosure Information

Robert J. Kreitman, MD
Senior Investigator
National Cancer Institute
National Institutes of Health
Bethesda, MD

Robert J. Kreitman, MD, has disclosed the following relevant financial relationships:

Coinventor on the NIH patent for Moxetumomab Pasudotox.

Receives research support and/or investigation drugs for trials from AstraZeneca, Pfizer, Novartis, Teva, and Genentech.

Dr. Robert J. Kreitman contributed to this article in his personal capacity. The views expressed are his own and do not necessarily represent tthe views of the National Institutes of Health or the United States Government.

Author and Disclosure Information

Robert J. Kreitman, MD
Senior Investigator
National Cancer Institute
National Institutes of Health
Bethesda, MD

Robert J. Kreitman, MD, has disclosed the following relevant financial relationships:

Coinventor on the NIH patent for Moxetumomab Pasudotox.

Receives research support and/or investigation drugs for trials from AstraZeneca, Pfizer, Novartis, Teva, and Genentech.

Dr. Robert J. Kreitman contributed to this article in his personal capacity. The views expressed are his own and do not necessarily represent tthe views of the National Institutes of Health or the United States Government.

Robert J. Kreitman, MD
Hairy cell leukemia (HCL) is an indolent, low-grade B-cell lymphoid malignancy that typically presents with fatigue, pancytopenia, and splenomegaly.1,2 It is a rare disease, with an estimated 1,200 new cases of HCL diagnosed annually in the United States.1 Demographically, HCL is a disease of older adults (median age at diagnosis, 58 years), and is more commonly found in men than women and in White individuals compared with other ethnic or racial backgrounds.3,4 Environmental or occupational exposure to toxic substances, pesticides, ionizing radiation, and petroleum products may be linked to increased risk for HCL development.1,4


Pathophysiology

HCL develops from activated, mature memory B-cells that, in most cases, have the acquired mutation in BRAF V600E, which is present in 80% to 90% of patients with classic HCL.1,3,5 BRAF is an integral part of the RAS-BRAF-MEK-ERK cellular pathway that transmits growth factor signals from the cell surface to the nucleus to regulate cell growth and proliferation.Mutated BRAF V600E continuously activates BRAF kinase and downstream signaling, resulting in enhanced HCL cell survival and unchecked proliferation.3

Variant HCL (HCLv) is a separate, more virulent disease that lacks BRAF V600E mutation and CD25 expression on flow cytometry.1,7-9 Patients with HCLv have a worse prognosis and poor responses to front-line purine analogs, and a higher proportion of these patients carry the unmutated immunoglobulin heavy chain variable (IGHV) gene (54% vs 17% in HCL).1,10,11 About 30% to 50% have wild-type BRAF and activating mutations in MAP2K1, which encodes aberrant MEK downstream of BRAF.10,12

Most patients with HCL have somatic mutations in the IGHV gene.3,13,14 Patients with unmutated IGHV4-34 and wildtype BRAF have an aggressive form of the disease, even if the HCL cells express CD25 as in classic HCL.1,15 HCL in patients with unmutated IGHV is often refractory to purine analogs and these patients have poor prognosis and rapid progression.16 Other identified mutations include CDKN1B in HCL and MAP2K1 and CCNC3 in HCLv.2

Signs and Symptoms

In many cases, HCL is asymptomatic, and diagnosed when pancytopenia, monocytopenia, and leukopenia are discovered on unrelated blood work.2,3,11 Monocytopenia is a specific presentation of HCL, but not HCLv.11 Typical systemic symptoms include unexplained weight loss and extreme fatigue (80%).1,3 Other symptoms can include fever, recurrent infections, night sweats, splenomegaly and related pain or abdominal fullness, hepatomegaly, and bleeding or bruising due to thrombocytopenia.1,3 Splenomegaly is associated with advanced disease.11

Up to 30% of patients may present with autoimmune disorders such as vasculitis or psoriasis. Although skin involvement is rare with HCL, 10% to 12% of patients will have dermatologic symptoms either due to recurrent infection or autoimmune reactions.1,2 Skin reactions include localized or generalized maculopapular rash, pyoderma gangrenosum (which may be severe), and recurrent bacterial or viral skin infections.17

Diagnosis

After complete history and physical examination, a diagnosis of HCL is usually made based on flow cytometry for immunophenotyping and molecular testing for BRAF V600E (Table 1).2,17

Table 1. Essential Tests for HCL Diagnosis

Disease-related fibrosis may impede bone marrow aspiration, and trephine biopsy should be done to make the diagnosis.11 On morphologic examination, HCL cells are small- to medium-sized, with round, oval, or indented, well-defined nuclei. Cytoplasm is pale blue, and cells have small cytoplasmic projections (Figure 1).2,18

©Getty Images/Kateryna Kon/Science Photo Library
Figure 1. Typical Appearance of Hairy Cell Leukemia

On flow cytometry, HCL is positive for B-cell antigens (CD19, CD20, CD22), as well as antigens specific to the disease (CD11c, CD25, CD103, CD123), and by immunohistochemistry (IHC) for cyclin D1 and annexin-A1. CD20, CD123, and CD200 are bright in HCL. The presence of T-cell marker CD103 on B-cells indicates HCL.1-3 HCLv, in contrast, is positive for CD11c and CD103, but usually negative for CD25, CD123, and annexin-A1.2,19

BRAF V600E mutation can be identified using droplet digital polymerase chain reaction (PCR), next-generation molecular sequencing, or IHC with a VE1 stain.3,11 IHC for CD20, annexin-1, and VE1 establish the diagnosis, but also are useful in determining the extent to which leukemic cells have infiltrated bone marrow.11

Differential diagnosis of HCL includes HCL variants, splenic marginal zone lymphoma, and splenic diffuse red pulp small B-cell lymphoma.7,11

Indications for Treatment and Criteria for Response

Over time, about 90% of patients with HCL will require treatment. However, not all such patients will require urgent or immediate treatment, and some can be managed with observation and close monitoring.1,11 The indications for initiating treatment generally are systemic symptoms and significant pancytopenia (Table 2).2,11

Table 3. Indications for Treatment of HCL

The optimal response with treatment of HCL is complete response (CR) without minimal residual disease (MRD-free), which minimizes the risk for relapse.1,11 Hematologic and molecular response is assessed using peripheral blood samples; physical examination, ultrasound, computed tomography, or magnetic resonance imaging is used to determine response in lymph nodes, spleen, or liver.1 MRD-free is defined by the absence of HCL cells by the chosen method (IHC, flow cytometry, or PCR).20 Bone marrow aspirate flow cytometry is the most sensitive standard test for MRD detection.1Table 3 summarizes response criteria for HCL.2,11

Table 3. Indications for Treatment of HCL

Initial Treatment of HCL

The purine nucleoside analogs (PNAs) cladribine (± rituximab) and pentostatin are widely recommended for initial treatment.1,2,11 As monotherapy, cladribine and pentostatin are considered similarly effective, with CR in 70% to 90% of patients and durations of response > 10 years.1 Adding the anti-CD20 monoclonal antibody rituximab in 8 weekly doses starting the first day of front-line cladribine (CDAR) improves remission, MRD-free rates, and duration of response (94% MRD-free at 96 months), with minimal added toxicity.21 Rituximab is often added 4 weeks after cladribine, which offers more convenience, an equally high CR rate of 100%, and a 76% MRD-free rate at 3 months.11 Bone marrow biopsy should be delayed for 4 to 6 months to allow a full response to develop with cladribine.1,11

Daily (intravenous or subcutaneous) and weekly cladribine are equally safe and effective.2,11 Pentostatin is administered intravenously every 2 weeks for 3 to 6 months, allowing time for hematologic recovery between doses.1,11 Patient factors to consider when choosing treatment include baseline neutropenia, patient preference, and comorbidities.

Toxicities of PNAs include neutropenia and fever, which typically occur during the first month of treatment and are more frequent in patients with baseline severe neutropenia; T-cell recovery may take years.1 CDAR is associated with higher transient thrombocytopenia, but faster platelet and neutrophil recovery at 4 weeks than cladribine alone.21 Both therapies are immunosuppressive. Patients should be evaluated for existing infections and watched for new infections during treatment. Control of active infection prior to treatment initiation is required.11,23

Patients with confirmed BRAF V600E mutation are candidates for vemurafenib if they are unable to tolerate a PNA, have an active infection, or would like effective vaccinations.2,23-25

Treatment at Relapse

At suspected HCL relapse, patients should be evaluated to determine whether cytopenia is due to recurrent disease or lingering effects from prior treatment. Use of successive flow cytometry over time can clarify whether symptoms are related to disease and need interventional treatment, or will resolve with additional time.1

Patients who have an HCL relapse after initial therapy with cladribine or pentostatin may be candidates for re-treatment with the same or alternate PNA plus rituximab.2 Rituximab
monotherapy has been used for patients unable to tolerate PNA but yields CR rates as low as 13%.26 Repeated courses of PNA therapy yield lower rates and durations of response with each course.1,2

For patients with primary refractory disease (less than CR with initial therapy) or relapse within 2 years of initial therapy, treatment with the BRAF V600E inhibitor vemurafenib off-label, with or without rituximab, is an option.2,5 In HCL, vemurafenib for patients with relapsed or refractory disease achieved CR in 35% and 42% in 2 small trials (N = 54). Relapse-free survival among people with CR was 19 months in 1 of the trials.27 Vemurafenib plus rituximab achieved CR in 87% of patients with relapsed or refractory HCL, and an MRD-free CR rate of 57%. Among patients with CR, 85% were relapse-free at a median follow-up of 34 months.5 Treatment with vemurafenib is not myelotoxic—an advantage for HCL patients. Adverse effects with vemurafenib are often manageable with dose reductions, if needed. A specific concern with vemurafenib is the potential development of secondary skin cancers.5,27,28

Novel Targeted Options and Recommended Use

Promising alternatives for patients with relapsed or refractory HCL include combined BRAF and MEK inhibitors and the Bruton tyrosine kinase (BTK) inhibitor ibrutinib. The concept of BRAF/MEK inhibition was validated in studies with BRAF-mutated melanoma, in which dabrafenib plus trametinib (the MEK inhibitor) improved overall survival (OS) with less toxicity and better quality of life than vemurafenib.1,29 In a phase 2 trial in HCL, dabrafenib monotherapy demonstrated an overall response rate (ORR) of 80%, including 30% CR.30 In a subsequent phase 2 trial, dabrafenib combined with trametinib was evaluated in refractory or late relapsed HCL. Among 55 enrolled patients, objective response rate was 89%, including 65.5% CR. Nine of 36 patients with CR were MRD-free. Among responding patients, duration of response was 97.7% at 24 months.31 The most common grade ≥ 3 toxicities were hyperglycemia, pyrexia, neutropenia, and pneumonia. Secondary skin cancers were seen in about 5% of patients.31

BRAF/MEK inhibitor combinations in HCL offer effective therapy with less myelosuppression than PNAs, making them useful for patients with or at risk for infection.23 Their use in HCL is off-label, as they currently are approved for treatment of BRAF-mutated melanoma and some other tumors.32 A study of encorafenib (a BRAF inhibitor) combined with binimetinib (a MEK inhibitor) is ongoing (Table 4).32

Table 4. Currently Recruiting Clinical Trials Specifically for HCL

Ibrutinib interrupts B-cell receptor signaling to stop tumor cell growth. In a phase 2 trial, patients with relapsed or refractory HCL or HCLv were treated with once-daily oral ibrutinib. Best ORR was 54% (19% CR; 3% MRD-free). Despite the low CR rate, 3-year progression-free survival with ibrutinib was 73% and OS was 85%. Treatment was well-tolerated; cytopenia (including 22% grade ≥ 3 thrombocytopenia and neutropenia) and diarrhea were frequent toxicities.33

Moxetumomab pasudotox is a novel CD22-targeted antibody fused with protein toxin that interrupts protein synthesis in tumor cells.1 As treatment, it was studied in a phase 3 trial of relapsed HCL in heavily pretreated patients, and achieved a CR rate of 41%, including 36% durable CR.34 Although FDA-approved for relapsed or refractory HCL, the drug is being discontinued due to business decisions, not safety or efficacy concerns.2 It is notable that many types of B-cell lymphoma also express CD22.35

Enrollment in a clinical trial to study possible treatment advances is recommended by the National Comprehensive Cancer Network (NCCN) at first and subsequent relapses of HCL for appropriate patients.Figure 2 summarizes an approach to treatment choice and sequencing for patients with HCL.

Figure 2. Standard & Investigational Treatment of HCL/HCLv

Supportive Care

Patients being treated for HCL should have supportive care to manage adverse effects of their disease. Such care includes prophylaxis against herpes virus if CD4+ T cells < 200 cells/μL and other prophylactic vaccinations to hepatitis B virus, COVID-19 and Influenza. Patients with neutropeni may require broad-spectrum antibacterial prophylaxis or neutrophil growth factors if neutropenic fever develops. Blood product support is recommended if needed.2 Assessment of anti-COVID-19 antibodies is recommended to optimize immunity, particularly prior to beginning anti-CD20 antibody therapy like rituximab.23

Unmet Needs

Despite improvements in response and survival with newer therapies, not all patients with HCL benefit from these advances. Unmet needs are finding optimal treatment for patients with HCLv, despite some success with MEK inhibitors, and for patients with BRAF mutations other than V600E, who have few options beyond PNAs and rituximab.

Click to read more from 2023 Rare Diseases Report: Cancers

Robert J. Kreitman, MD
Hairy cell leukemia (HCL) is an indolent, low-grade B-cell lymphoid malignancy that typically presents with fatigue, pancytopenia, and splenomegaly.1,2 It is a rare disease, with an estimated 1,200 new cases of HCL diagnosed annually in the United States.1 Demographically, HCL is a disease of older adults (median age at diagnosis, 58 years), and is more commonly found in men than women and in White individuals compared with other ethnic or racial backgrounds.3,4 Environmental or occupational exposure to toxic substances, pesticides, ionizing radiation, and petroleum products may be linked to increased risk for HCL development.1,4


Pathophysiology

HCL develops from activated, mature memory B-cells that, in most cases, have the acquired mutation in BRAF V600E, which is present in 80% to 90% of patients with classic HCL.1,3,5 BRAF is an integral part of the RAS-BRAF-MEK-ERK cellular pathway that transmits growth factor signals from the cell surface to the nucleus to regulate cell growth and proliferation.Mutated BRAF V600E continuously activates BRAF kinase and downstream signaling, resulting in enhanced HCL cell survival and unchecked proliferation.3

Variant HCL (HCLv) is a separate, more virulent disease that lacks BRAF V600E mutation and CD25 expression on flow cytometry.1,7-9 Patients with HCLv have a worse prognosis and poor responses to front-line purine analogs, and a higher proportion of these patients carry the unmutated immunoglobulin heavy chain variable (IGHV) gene (54% vs 17% in HCL).1,10,11 About 30% to 50% have wild-type BRAF and activating mutations in MAP2K1, which encodes aberrant MEK downstream of BRAF.10,12

Most patients with HCL have somatic mutations in the IGHV gene.3,13,14 Patients with unmutated IGHV4-34 and wildtype BRAF have an aggressive form of the disease, even if the HCL cells express CD25 as in classic HCL.1,15 HCL in patients with unmutated IGHV is often refractory to purine analogs and these patients have poor prognosis and rapid progression.16 Other identified mutations include CDKN1B in HCL and MAP2K1 and CCNC3 in HCLv.2

Signs and Symptoms

In many cases, HCL is asymptomatic, and diagnosed when pancytopenia, monocytopenia, and leukopenia are discovered on unrelated blood work.2,3,11 Monocytopenia is a specific presentation of HCL, but not HCLv.11 Typical systemic symptoms include unexplained weight loss and extreme fatigue (80%).1,3 Other symptoms can include fever, recurrent infections, night sweats, splenomegaly and related pain or abdominal fullness, hepatomegaly, and bleeding or bruising due to thrombocytopenia.1,3 Splenomegaly is associated with advanced disease.11

Up to 30% of patients may present with autoimmune disorders such as vasculitis or psoriasis. Although skin involvement is rare with HCL, 10% to 12% of patients will have dermatologic symptoms either due to recurrent infection or autoimmune reactions.1,2 Skin reactions include localized or generalized maculopapular rash, pyoderma gangrenosum (which may be severe), and recurrent bacterial or viral skin infections.17

Diagnosis

After complete history and physical examination, a diagnosis of HCL is usually made based on flow cytometry for immunophenotyping and molecular testing for BRAF V600E (Table 1).2,17

Table 1. Essential Tests for HCL Diagnosis

Disease-related fibrosis may impede bone marrow aspiration, and trephine biopsy should be done to make the diagnosis.11 On morphologic examination, HCL cells are small- to medium-sized, with round, oval, or indented, well-defined nuclei. Cytoplasm is pale blue, and cells have small cytoplasmic projections (Figure 1).2,18

©Getty Images/Kateryna Kon/Science Photo Library
Figure 1. Typical Appearance of Hairy Cell Leukemia

On flow cytometry, HCL is positive for B-cell antigens (CD19, CD20, CD22), as well as antigens specific to the disease (CD11c, CD25, CD103, CD123), and by immunohistochemistry (IHC) for cyclin D1 and annexin-A1. CD20, CD123, and CD200 are bright in HCL. The presence of T-cell marker CD103 on B-cells indicates HCL.1-3 HCLv, in contrast, is positive for CD11c and CD103, but usually negative for CD25, CD123, and annexin-A1.2,19

BRAF V600E mutation can be identified using droplet digital polymerase chain reaction (PCR), next-generation molecular sequencing, or IHC with a VE1 stain.3,11 IHC for CD20, annexin-1, and VE1 establish the diagnosis, but also are useful in determining the extent to which leukemic cells have infiltrated bone marrow.11

Differential diagnosis of HCL includes HCL variants, splenic marginal zone lymphoma, and splenic diffuse red pulp small B-cell lymphoma.7,11

Indications for Treatment and Criteria for Response

Over time, about 90% of patients with HCL will require treatment. However, not all such patients will require urgent or immediate treatment, and some can be managed with observation and close monitoring.1,11 The indications for initiating treatment generally are systemic symptoms and significant pancytopenia (Table 2).2,11

Table 3. Indications for Treatment of HCL

The optimal response with treatment of HCL is complete response (CR) without minimal residual disease (MRD-free), which minimizes the risk for relapse.1,11 Hematologic and molecular response is assessed using peripheral blood samples; physical examination, ultrasound, computed tomography, or magnetic resonance imaging is used to determine response in lymph nodes, spleen, or liver.1 MRD-free is defined by the absence of HCL cells by the chosen method (IHC, flow cytometry, or PCR).20 Bone marrow aspirate flow cytometry is the most sensitive standard test for MRD detection.1Table 3 summarizes response criteria for HCL.2,11

Table 3. Indications for Treatment of HCL

Initial Treatment of HCL

The purine nucleoside analogs (PNAs) cladribine (± rituximab) and pentostatin are widely recommended for initial treatment.1,2,11 As monotherapy, cladribine and pentostatin are considered similarly effective, with CR in 70% to 90% of patients and durations of response > 10 years.1 Adding the anti-CD20 monoclonal antibody rituximab in 8 weekly doses starting the first day of front-line cladribine (CDAR) improves remission, MRD-free rates, and duration of response (94% MRD-free at 96 months), with minimal added toxicity.21 Rituximab is often added 4 weeks after cladribine, which offers more convenience, an equally high CR rate of 100%, and a 76% MRD-free rate at 3 months.11 Bone marrow biopsy should be delayed for 4 to 6 months to allow a full response to develop with cladribine.1,11

Daily (intravenous or subcutaneous) and weekly cladribine are equally safe and effective.2,11 Pentostatin is administered intravenously every 2 weeks for 3 to 6 months, allowing time for hematologic recovery between doses.1,11 Patient factors to consider when choosing treatment include baseline neutropenia, patient preference, and comorbidities.

Toxicities of PNAs include neutropenia and fever, which typically occur during the first month of treatment and are more frequent in patients with baseline severe neutropenia; T-cell recovery may take years.1 CDAR is associated with higher transient thrombocytopenia, but faster platelet and neutrophil recovery at 4 weeks than cladribine alone.21 Both therapies are immunosuppressive. Patients should be evaluated for existing infections and watched for new infections during treatment. Control of active infection prior to treatment initiation is required.11,23

Patients with confirmed BRAF V600E mutation are candidates for vemurafenib if they are unable to tolerate a PNA, have an active infection, or would like effective vaccinations.2,23-25

Treatment at Relapse

At suspected HCL relapse, patients should be evaluated to determine whether cytopenia is due to recurrent disease or lingering effects from prior treatment. Use of successive flow cytometry over time can clarify whether symptoms are related to disease and need interventional treatment, or will resolve with additional time.1

Patients who have an HCL relapse after initial therapy with cladribine or pentostatin may be candidates for re-treatment with the same or alternate PNA plus rituximab.2 Rituximab
monotherapy has been used for patients unable to tolerate PNA but yields CR rates as low as 13%.26 Repeated courses of PNA therapy yield lower rates and durations of response with each course.1,2

For patients with primary refractory disease (less than CR with initial therapy) or relapse within 2 years of initial therapy, treatment with the BRAF V600E inhibitor vemurafenib off-label, with or without rituximab, is an option.2,5 In HCL, vemurafenib for patients with relapsed or refractory disease achieved CR in 35% and 42% in 2 small trials (N = 54). Relapse-free survival among people with CR was 19 months in 1 of the trials.27 Vemurafenib plus rituximab achieved CR in 87% of patients with relapsed or refractory HCL, and an MRD-free CR rate of 57%. Among patients with CR, 85% were relapse-free at a median follow-up of 34 months.5 Treatment with vemurafenib is not myelotoxic—an advantage for HCL patients. Adverse effects with vemurafenib are often manageable with dose reductions, if needed. A specific concern with vemurafenib is the potential development of secondary skin cancers.5,27,28

Novel Targeted Options and Recommended Use

Promising alternatives for patients with relapsed or refractory HCL include combined BRAF and MEK inhibitors and the Bruton tyrosine kinase (BTK) inhibitor ibrutinib. The concept of BRAF/MEK inhibition was validated in studies with BRAF-mutated melanoma, in which dabrafenib plus trametinib (the MEK inhibitor) improved overall survival (OS) with less toxicity and better quality of life than vemurafenib.1,29 In a phase 2 trial in HCL, dabrafenib monotherapy demonstrated an overall response rate (ORR) of 80%, including 30% CR.30 In a subsequent phase 2 trial, dabrafenib combined with trametinib was evaluated in refractory or late relapsed HCL. Among 55 enrolled patients, objective response rate was 89%, including 65.5% CR. Nine of 36 patients with CR were MRD-free. Among responding patients, duration of response was 97.7% at 24 months.31 The most common grade ≥ 3 toxicities were hyperglycemia, pyrexia, neutropenia, and pneumonia. Secondary skin cancers were seen in about 5% of patients.31

BRAF/MEK inhibitor combinations in HCL offer effective therapy with less myelosuppression than PNAs, making them useful for patients with or at risk for infection.23 Their use in HCL is off-label, as they currently are approved for treatment of BRAF-mutated melanoma and some other tumors.32 A study of encorafenib (a BRAF inhibitor) combined with binimetinib (a MEK inhibitor) is ongoing (Table 4).32

Table 4. Currently Recruiting Clinical Trials Specifically for HCL

Ibrutinib interrupts B-cell receptor signaling to stop tumor cell growth. In a phase 2 trial, patients with relapsed or refractory HCL or HCLv were treated with once-daily oral ibrutinib. Best ORR was 54% (19% CR; 3% MRD-free). Despite the low CR rate, 3-year progression-free survival with ibrutinib was 73% and OS was 85%. Treatment was well-tolerated; cytopenia (including 22% grade ≥ 3 thrombocytopenia and neutropenia) and diarrhea were frequent toxicities.33

Moxetumomab pasudotox is a novel CD22-targeted antibody fused with protein toxin that interrupts protein synthesis in tumor cells.1 As treatment, it was studied in a phase 3 trial of relapsed HCL in heavily pretreated patients, and achieved a CR rate of 41%, including 36% durable CR.34 Although FDA-approved for relapsed or refractory HCL, the drug is being discontinued due to business decisions, not safety or efficacy concerns.2 It is notable that many types of B-cell lymphoma also express CD22.35

Enrollment in a clinical trial to study possible treatment advances is recommended by the National Comprehensive Cancer Network (NCCN) at first and subsequent relapses of HCL for appropriate patients.Figure 2 summarizes an approach to treatment choice and sequencing for patients with HCL.

Figure 2. Standard & Investigational Treatment of HCL/HCLv

Supportive Care

Patients being treated for HCL should have supportive care to manage adverse effects of their disease. Such care includes prophylaxis against herpes virus if CD4+ T cells < 200 cells/μL and other prophylactic vaccinations to hepatitis B virus, COVID-19 and Influenza. Patients with neutropeni may require broad-spectrum antibacterial prophylaxis or neutrophil growth factors if neutropenic fever develops. Blood product support is recommended if needed.2 Assessment of anti-COVID-19 antibodies is recommended to optimize immunity, particularly prior to beginning anti-CD20 antibody therapy like rituximab.23

Unmet Needs

Despite improvements in response and survival with newer therapies, not all patients with HCL benefit from these advances. Unmet needs are finding optimal treatment for patients with HCLv, despite some success with MEK inhibitors, and for patients with BRAF mutations other than V600E, who have few options beyond PNAs and rituximab.

Click to read more from 2023 Rare Diseases Report: Cancers

References
  1. Kreitman RJ, Arons E. Diagnosis and treatment of hairy cell leukemia as the COVID-19 pandemic continues. Blood Rev. 2022;51:100888. doi:10.1016/j.blre.2021.100888
  2. National Comprehensive Cancer Network. NCCN clinical practice guideline in oncology: hairy cell leukemia. Version 1.2023. Published August 30, 2022. Accessed March 16, 2023. https://www.nccn.org/professionals/physician_gls/pdf/hairy_cell.pdf
  3. Janus A, Robak T. Hairy cell leukemia. In: Li W, ed. Leukemia [Internet]. Brisbane: Exon Publications; 2022:chap3. Accessed February 16, 2023. doi:10.36255/exon-publications-leukemia-hairy-cell-leukemia
  4. Tadmor T, Polliack A. Epidemiology and environmental risk in hairy cell leukemia. Best Pract Res Clin Haematol. 2015;28(4):175-179. doi:10.1016/j.beha.2015.10.014
  5. Tiacci E, De Carolis L, Simonetti E, et al. Vemurafenib plus rituximab in refractory or relapsed hairy-cell leukemia. N Engl J Med. 2021;384(19):1810-1823. doi:10.1056/NEJMoa20312986
  6. Falini B, Martelli MP, Tiacci E. BRAF V600E mutation in hairy cell leukemia: from bench to bedside. Blood. 2016;128(15):1918-1927. doi:10.1182/blood-2016-07-418434
  7. Matutes E. Diagnostic and therapeutic challenges in hairy cell leukemia-variant: where are we in 2021? Expert Rev Hematol. 2021;14(4):355-363. doi:10.1080/17474086.2021.1908121
  8. Cawley JC, Burns GF, Hayhoe FG. A chronic lymphoproliferative disorder with distinctive features: a distinct variant of hairy-cell leukaemia. Leuk Res. 1980;4(6):547-559. doi:10.1016/0145-2126(80)90066-1
  9. Xi L, Arons E, Navarro W, et al. Both variant and IGHV4-34-expressing hairy cell leukemia lack the BRAF V600E mutation. Blood. 2012;119(14):3330-3332. doi:10.1182/blood-2011-09-379339
  10. Durham BH, Getta B, Dietrich S, et al. Genomic analysis of hairy cell leukemia identifies novel recurrent genetic alterations. Blood. 2017;130(14):1644-1648. doi:10.1182/blood-2017-01-76510711
  11. Grever MR, Abdel-Wahab O, Andritsos LA, et al. Consensus guidelines for the diagnosis and management of patients with hairy cell leukemia. Blood. 2017;129(5):553-560. doi:10.1182/blood-2016-01-689422
  12. Waterfall JJ, Arons E, Walker RL, et al. High prevalence of MAP2K1 mutations in variant and IGHV4-34-expressing hairy-cell leukemias. Nat Genet. 2014;46(1):8-10. doi:10.1038/ng.2828
  13. Arons E, Sunshine J, Suntum T, Kreitman RJ. Somatic hypermutation and VH gene usage in hairy cell leukaemia. Br J Haematol. 2006;133(5):504-512. doi:10.1111/j.1365-2141.2006.06066.x
  14. Arons E, Roth L, Sapolsky J, Suntum T, Stetler-Stevenson M, Kreitman RJ. Evidence of canonical somatic hypermutation in hairy cell leukemia. Blood. 2011;117(18):4844-4851. doi:10.1182/blood-2010-11-316737
  15. Arons E, Suntum T, Stetler-Stevenson M, Kreitman RJ. VH4-34+ hairy cell leukemia, a new variant with poor prognosis despite standard therapy. Blood. 2009;114(21):4687-4695. doi:10.1182/blood-2009-01-201731
  16. Forconi F, Sozzi E, Cencini E, et al. Hairy cell leukemias with unmutated IGHV genes define the minor subset refractory to single-agent cladribine and with more aggressive behavior. Blood. 2009;114(21):4696-4702. doi:10.1182/blood-2009-03-212449
  17. Robak E, Jesionek-Kupnicka D, Robak T. Skin changes in hairy cell leukemia. Ann Hematol. 2021;100(3):615-625. doi:10.1007/s00277-020-04349-z
  18. Bouroncle BA. Thirty-five years in the progress of hairy cell leukemia. Leuk Lymphoma. 1994;14(suppl 1):1-12. https://pubmed.ncbi.nlm.nih.gov/7820038/
  19. Falini B, Tiacci E, Liso A, et al. Simple diagnostic assay for hairy cell leukaemia by immunocytochemical detection of annexin A1 (ANXA1). Lancet. 2004;363(9424): 1869-1870. doi:10.1016/S0140-6736(04)16356-3
  20. Robak T, Robak P. Measurable residual disease in hairy cell leukemia: technical considerations and clinical significance. Front Oncol. 2022;12:976374. doi:10.3389/fonc.2022.976374
  21. Chihara D, Arons E, Stetler-Stevenson M, et al. Randomized phase II study of first-line cladribine with concurrent or delayed rituximab in patients with hairy cell leukemia. J Clin Oncol. 2020;38(14):1527-1538. doi:10.1200/JCO.19.02250
  22. Chihara D, Kantarjian H, O’Brien S, et al. Long-term durable remission by cladribine followed by rituximab in patients with hairy cell leukaemia: update of a phase II trial. Br J Haematol. 2016;174(5):760-766. doi:10.1111/bjh.14129
  23. Grever M, Andritsos L, Banerji V, et al. Hairy cell leukemia and COVID-19 adaptation of treatment guidelines. Leukemia. 2021;35(7):1864-1872. doi:10.1038/s41375-021-01257-7
  24. Konrat J, Rösler W, Roiss M, et al. BRAF inhibitor treatment of classical hairy cell leukemia allows successful vaccination against SARS-CoV-2. Ann Hematol. 2023;102(2):403-406. doi:10.1007/s00277-022-05026-z
  25. Park JH, Shukla M, Salcedo JM, et al. First-line chemo-free therapy with the BRAF inhibitor vemurafenib combined with obinutuzumab is effective in patients with HCL. Blood. 2019;134(suppl 1):Abstract 3998. https://doi.org/10.1182/blood-2019-124478
  26. Nieva J, Bethel K, Saven A. Phase 2 study of rituximab in the treatment of cladribine-failed patients with hairy cell leukemia. Blood. 2003;102(3):810-813. doi:10.1182/blood-2003-01-0014
  27. Tiacci E, Park JH, De Carolis L, et al. Targeting mutant BRAF in relapsed or refractory hairy-cell leukemia. N Engl J Med. 2015;373(18):1733-1747. doi:10.1056/NEJMoa1506583
  28. Maitre E, Paillassa J, Troussard X. Novel targeted treatments in hairy cell leukemia and other hairy cell-like disorders. Front Oncol. 2022;12:1068981. doi:10.3389/fonc.2022.1068981
  29. Grob JJ, Amonkar MM, Karaszewska B, et al. Comparison of dabrafenib and trametinib combination therapy with vemurafenib monotherapy on health-related quality of life in patients with unresectable or metastatic cutaneous BRAF Val600-mutation-positive melanoma (COMBI-v): results of a phase 3, open-label, randomised trial. Lancet Oncol. 2015;16(13):1389-1398. doi:10.1016/S1470-2045(15)00087-X
  30. Tiacci E, De Carolis L, Simonetti E, et al. Safety and efficacy of the BRAF inhibitor dabrafenib in relapsed or refractory hairy cell leukemia: a pilot phase-2 clinical trial. Leukemia. 2021;35(11):3314-3318. doi:10.1038/s41375-021-01210-8
  31. Kreitman RJ, Moreau P, Ravandi F, et al. Dabrafenib plus trametinib in patients with relapsed/refractory BRAF V600E mutation-positive hairy cell leukemia. Blood. 2023;141(9):996-1006. doi:10.1182/blood.2021013658
  32. Adashek JJ, Menta AK, Reddy NK, Desai AP, Roszik J, Subbiah V. Tissue agnostic activity of BRAF plus MEK inhibitor in BRAF V600E-mutated tumors. Mol Cancer Ther. 2022;21(6):871-878. doi:10.1158/1535-7163.MCT-21-0950
  33. Rogers KA, Andritsos LA, Wei L, et al. Phase 2 study of ibrutinib in classic and variant hairy cell leukemia. Blood. 2021;137(25):3473-3483. doi:10.1182/blood.2020009688
  34. Kreitman RJ, Dearden C, Zinzani PL, et al; Study 1053 investigators. Moxetumomab pasudotox in heavily pre-treated patients with relapsed/refractory hairy cell leukemia (HCL): long-term follow-up from the pivotal trial. J Hematol Oncol. 2021;14(1):35. doi:10.1186/s13045-020-01004-y
  35. Leonard JP, Goldenberg DM. Preclinical and clinical evaluation of epratuzumab (anti-CD22 IgG) in B-cell malignancies. Oncogene. 2007;26(25):3704-3713. doi:10.1038/sj.onc.1210370
References
  1. Kreitman RJ, Arons E. Diagnosis and treatment of hairy cell leukemia as the COVID-19 pandemic continues. Blood Rev. 2022;51:100888. doi:10.1016/j.blre.2021.100888
  2. National Comprehensive Cancer Network. NCCN clinical practice guideline in oncology: hairy cell leukemia. Version 1.2023. Published August 30, 2022. Accessed March 16, 2023. https://www.nccn.org/professionals/physician_gls/pdf/hairy_cell.pdf
  3. Janus A, Robak T. Hairy cell leukemia. In: Li W, ed. Leukemia [Internet]. Brisbane: Exon Publications; 2022:chap3. Accessed February 16, 2023. doi:10.36255/exon-publications-leukemia-hairy-cell-leukemia
  4. Tadmor T, Polliack A. Epidemiology and environmental risk in hairy cell leukemia. Best Pract Res Clin Haematol. 2015;28(4):175-179. doi:10.1016/j.beha.2015.10.014
  5. Tiacci E, De Carolis L, Simonetti E, et al. Vemurafenib plus rituximab in refractory or relapsed hairy-cell leukemia. N Engl J Med. 2021;384(19):1810-1823. doi:10.1056/NEJMoa20312986
  6. Falini B, Martelli MP, Tiacci E. BRAF V600E mutation in hairy cell leukemia: from bench to bedside. Blood. 2016;128(15):1918-1927. doi:10.1182/blood-2016-07-418434
  7. Matutes E. Diagnostic and therapeutic challenges in hairy cell leukemia-variant: where are we in 2021? Expert Rev Hematol. 2021;14(4):355-363. doi:10.1080/17474086.2021.1908121
  8. Cawley JC, Burns GF, Hayhoe FG. A chronic lymphoproliferative disorder with distinctive features: a distinct variant of hairy-cell leukaemia. Leuk Res. 1980;4(6):547-559. doi:10.1016/0145-2126(80)90066-1
  9. Xi L, Arons E, Navarro W, et al. Both variant and IGHV4-34-expressing hairy cell leukemia lack the BRAF V600E mutation. Blood. 2012;119(14):3330-3332. doi:10.1182/blood-2011-09-379339
  10. Durham BH, Getta B, Dietrich S, et al. Genomic analysis of hairy cell leukemia identifies novel recurrent genetic alterations. Blood. 2017;130(14):1644-1648. doi:10.1182/blood-2017-01-76510711
  11. Grever MR, Abdel-Wahab O, Andritsos LA, et al. Consensus guidelines for the diagnosis and management of patients with hairy cell leukemia. Blood. 2017;129(5):553-560. doi:10.1182/blood-2016-01-689422
  12. Waterfall JJ, Arons E, Walker RL, et al. High prevalence of MAP2K1 mutations in variant and IGHV4-34-expressing hairy-cell leukemias. Nat Genet. 2014;46(1):8-10. doi:10.1038/ng.2828
  13. Arons E, Sunshine J, Suntum T, Kreitman RJ. Somatic hypermutation and VH gene usage in hairy cell leukaemia. Br J Haematol. 2006;133(5):504-512. doi:10.1111/j.1365-2141.2006.06066.x
  14. Arons E, Roth L, Sapolsky J, Suntum T, Stetler-Stevenson M, Kreitman RJ. Evidence of canonical somatic hypermutation in hairy cell leukemia. Blood. 2011;117(18):4844-4851. doi:10.1182/blood-2010-11-316737
  15. Arons E, Suntum T, Stetler-Stevenson M, Kreitman RJ. VH4-34+ hairy cell leukemia, a new variant with poor prognosis despite standard therapy. Blood. 2009;114(21):4687-4695. doi:10.1182/blood-2009-01-201731
  16. Forconi F, Sozzi E, Cencini E, et al. Hairy cell leukemias with unmutated IGHV genes define the minor subset refractory to single-agent cladribine and with more aggressive behavior. Blood. 2009;114(21):4696-4702. doi:10.1182/blood-2009-03-212449
  17. Robak E, Jesionek-Kupnicka D, Robak T. Skin changes in hairy cell leukemia. Ann Hematol. 2021;100(3):615-625. doi:10.1007/s00277-020-04349-z
  18. Bouroncle BA. Thirty-five years in the progress of hairy cell leukemia. Leuk Lymphoma. 1994;14(suppl 1):1-12. https://pubmed.ncbi.nlm.nih.gov/7820038/
  19. Falini B, Tiacci E, Liso A, et al. Simple diagnostic assay for hairy cell leukaemia by immunocytochemical detection of annexin A1 (ANXA1). Lancet. 2004;363(9424): 1869-1870. doi:10.1016/S0140-6736(04)16356-3
  20. Robak T, Robak P. Measurable residual disease in hairy cell leukemia: technical considerations and clinical significance. Front Oncol. 2022;12:976374. doi:10.3389/fonc.2022.976374
  21. Chihara D, Arons E, Stetler-Stevenson M, et al. Randomized phase II study of first-line cladribine with concurrent or delayed rituximab in patients with hairy cell leukemia. J Clin Oncol. 2020;38(14):1527-1538. doi:10.1200/JCO.19.02250
  22. Chihara D, Kantarjian H, O’Brien S, et al. Long-term durable remission by cladribine followed by rituximab in patients with hairy cell leukaemia: update of a phase II trial. Br J Haematol. 2016;174(5):760-766. doi:10.1111/bjh.14129
  23. Grever M, Andritsos L, Banerji V, et al. Hairy cell leukemia and COVID-19 adaptation of treatment guidelines. Leukemia. 2021;35(7):1864-1872. doi:10.1038/s41375-021-01257-7
  24. Konrat J, Rösler W, Roiss M, et al. BRAF inhibitor treatment of classical hairy cell leukemia allows successful vaccination against SARS-CoV-2. Ann Hematol. 2023;102(2):403-406. doi:10.1007/s00277-022-05026-z
  25. Park JH, Shukla M, Salcedo JM, et al. First-line chemo-free therapy with the BRAF inhibitor vemurafenib combined with obinutuzumab is effective in patients with HCL. Blood. 2019;134(suppl 1):Abstract 3998. https://doi.org/10.1182/blood-2019-124478
  26. Nieva J, Bethel K, Saven A. Phase 2 study of rituximab in the treatment of cladribine-failed patients with hairy cell leukemia. Blood. 2003;102(3):810-813. doi:10.1182/blood-2003-01-0014
  27. Tiacci E, Park JH, De Carolis L, et al. Targeting mutant BRAF in relapsed or refractory hairy-cell leukemia. N Engl J Med. 2015;373(18):1733-1747. doi:10.1056/NEJMoa1506583
  28. Maitre E, Paillassa J, Troussard X. Novel targeted treatments in hairy cell leukemia and other hairy cell-like disorders. Front Oncol. 2022;12:1068981. doi:10.3389/fonc.2022.1068981
  29. Grob JJ, Amonkar MM, Karaszewska B, et al. Comparison of dabrafenib and trametinib combination therapy with vemurafenib monotherapy on health-related quality of life in patients with unresectable or metastatic cutaneous BRAF Val600-mutation-positive melanoma (COMBI-v): results of a phase 3, open-label, randomised trial. Lancet Oncol. 2015;16(13):1389-1398. doi:10.1016/S1470-2045(15)00087-X
  30. Tiacci E, De Carolis L, Simonetti E, et al. Safety and efficacy of the BRAF inhibitor dabrafenib in relapsed or refractory hairy cell leukemia: a pilot phase-2 clinical trial. Leukemia. 2021;35(11):3314-3318. doi:10.1038/s41375-021-01210-8
  31. Kreitman RJ, Moreau P, Ravandi F, et al. Dabrafenib plus trametinib in patients with relapsed/refractory BRAF V600E mutation-positive hairy cell leukemia. Blood. 2023;141(9):996-1006. doi:10.1182/blood.2021013658
  32. Adashek JJ, Menta AK, Reddy NK, Desai AP, Roszik J, Subbiah V. Tissue agnostic activity of BRAF plus MEK inhibitor in BRAF V600E-mutated tumors. Mol Cancer Ther. 2022;21(6):871-878. doi:10.1158/1535-7163.MCT-21-0950
  33. Rogers KA, Andritsos LA, Wei L, et al. Phase 2 study of ibrutinib in classic and variant hairy cell leukemia. Blood. 2021;137(25):3473-3483. doi:10.1182/blood.2020009688
  34. Kreitman RJ, Dearden C, Zinzani PL, et al; Study 1053 investigators. Moxetumomab pasudotox in heavily pre-treated patients with relapsed/refractory hairy cell leukemia (HCL): long-term follow-up from the pivotal trial. J Hematol Oncol. 2021;14(1):35. doi:10.1186/s13045-020-01004-y
  35. Leonard JP, Goldenberg DM. Preclinical and clinical evaluation of epratuzumab (anti-CD22 IgG) in B-cell malignancies. Oncogene. 2007;26(25):3704-3713. doi:10.1038/sj.onc.1210370
Publications
Publications
Topics
Article Type
Display Headline
Advances in Management of Relapsed/Refractory Hairy Cell Leukemia
Display Headline
Advances in Management of Relapsed/Refractory Hairy Cell Leukemia
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Thu, 06/22/2023 - 18:45
Un-Gate On Date
Thu, 06/22/2023 - 18:45
Use ProPublica
CFC Schedule Remove Status
Thu, 06/22/2023 - 18:45
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Progress in Treating Testicular Cancer

Article Type
Changed
Mon, 06/26/2023 - 12:26
Display Headline
Progress in Treating Testicular Cancer

Liang Cheng, MD
Approximately 1% of adult neoplasms and 5% of all urologic cancers are testicular cancer (TC).1 In the United States, 9190 new cases have been estimated for 2023.1 Testicular germ cell tumors (GCTs) comprise 90% to 95% of all TCs and are grouped into seminomas, nonseminomatous GCTs (NSGCTs), and mixed histology GCTs.1 NSGCTs tend to be more aggressive and are more common in younger men (15-40 years old), whereas seminomas are slower growing and generally develop later in a patient’s life.2,3 


Mortality from TC has been decreasing since the 1970s due to cisplatin-based chemotherapy regimens2,3; TC is among the most curable of solid neoplasms, with a 5-year relative survival rate of 95%.2-4 Thus, the focus of research has shifted from optimizing treatments for improved survival to decreasing treatment-related, long-term adverse events (AEs).5

New Modifications in Risk Assessment and Prognostication

The widely accepted risk stratification model in use today was first developed in 1997 by the International Germ Cell Cancer Collaborative Group (IGCCCG) after studying data on patients with seminoma and NSGCTs.6 The original classification categorized metastatic NSGCTs as having good, intermediate, or poor prognosis based on levels of alpha-fetoprotein (AFP), human chorionic gonadotropin (hCG), lactate dehydrogenase (LDH), and the presence of nonpulmonary visceral metastases (NPVM). Primary mediastinal NSGCTs were classified as having poor prognosis regardless of the other factors.Metastatic seminoma GCTs were categorized as having good or intermediate prognosis based on the occurrence of brain, liver, or bone metastasis.7

Using contemporary data from more than 12,000 patients with metastatic GCTs who received either cisplatin or etoposide, the IGCCCG model was updated in 2021. For seminoma GCTs, 5-year progression-free survival (PFS) and 5-year overall survival (OS) were extended for both good and intermediate prognostic groups.7 LDH remained the most significant prognostic factor for determining good prognosis however, patients with LDH above 2.5× upper limit of normal (ULN) before chemotherapy had worse survival probabilities than patients with LDH at 2.5× ULN or lower. The survival probabilities for patients with otherwise good prognosis with LDH of more than 2.5× ULN were like those for patients with intermediate prognosis.7 Thus, using LDH of more than 2.5× ULN has revealed a subgroup with significantly worse outcomes within the “good” prognostic group.7,8

For NSGCTs, 5-year PFS rates did not differ from the original IGCCCG for good and intermediate prognostic groups; however, the 2021 update revealed an improved PFS for the poor prognostic group. The 2021 update also demonstrated that 5-year OS rates improved for each group, and further confirmed that the 2 most important prognostic factors for NSGCT were the presence of NPVM and the presence of a mediastinal primary tumor. The update added 2 new adverse prognostic variables: age and metastases. Risk of progression increases 25% with every decade-of-life increase, and 66% with the presence of lung metastases. The LDH groups were reduced to a single cutoff at 2.5× ULN for NSGCTs.8

Primary and Subsequent Treatments for TC

Guideline-directed first-line and subsequent treatments for seminomas and NSGCTs have been developed by several organizations, including the National Comprehensive Cancer Network, IGCCCG, and the American Urological Association (see Figure 1 and 2). An analysis of the most used treatments was performed using the National Cancer Database.2 Most patients underwent orchiectomy without chemotherapy or radiation for both stage I seminomas (78%) and NSGCTs (57%). For stage II and III seminomas, most patients underwent surgery with chemotherapy (66% and 68%, respectively). Nearly half of patients with stage II NSGCTs were treated with surgery and chemotherapy (49%), and a third were treated with retroperitoneal lymph node dissection (RPLND) in addition to surgery and chemotherapy. Surgery with chemotherapy was used for 55% of stage III NSGCTs; other treatments included surgery combined with chemotherapy and RPLND (19%), and chemotherapy with or without radiation (20%).2 However, nearly 30% of patients with TC do not receive guideline-directed therapy, including inappropriate imaging and overtreatment; and nonguideline–directed therapy has been independently associated with risk of relapse.12,13

Figure 1. First-line and Subsequent Treatment of Seminomas

Figure 2. First-line and Subsequent Treatment of Non-Seminomas

TC Survivorship

The trend of improved OS after treatment for metastatic GCTs highlights a need to focus on survivorship. The 10-year survival rate for TC post-treatment is 95%.14 Latest estimates suggest there are more than 300,000 TC survivors in the United States,accounting for approximately 4% of all US male cancer survivors.14 With longer-term survival, however, comes the risk for long-term complications from cancer treatments. For example, circulating platinum has been detected in the plasma of men up to 28 years after undergoing cisplatin-based chemotherapy for TC.15 Increasing levels of residual serum platinum have also been shown to correlate with severity of neurotoxicity between 5 and 20 years after treatment.16

A significant concern with cancer treatment is the development of second malignant neoplasms (SMNs).14,17 The relative risk of the development of SMNs depends on whetherradiation therapy or chemotherapy, or both, was used as the primary treatment. Patients who received either radiation therapy or chemotherapy are at increased risk for leukemia and solid cancers, including gastrointestinal cancers. For patients treated with cisplatin, a significant dose-response relationship between cumulative dose and leukemic risk has been reported.14

Other concerns are increased non-TC mortality and SMN mortality. Hellesnes et al examined cause-specific, non-TC mortality using a population-based cohort in Norway.18 They determined that the overall 25-year, non-TC mortality risk was 13.7% (95% CI, 12.5-14.9) for patients who previously had TC vs 11.3% for patients who never had TC. The highest mortality rates were reported for patients who had radiation (19%) or platinum-based chemotherapy plus radiation (18.4%); the lowest mortality rate was reported for patients who had received platinum-based chemotherapy only (9.5%). Patients with the highest non-TC mortality risk were fewer than 20 years post-cancer diagnosis. Non-TC mortality excess ranged from 23% to 40% for patients with a prior TC diagnosis, and a significant 1.43- to 3.24-fold increase in SMN mortality emerged after treatment with platinum-based chemotherapy or radiation therapy, or both.19 Awareness of the increased premature mortality risk is crucial for both TC survivors and their care providers.18

Quality of life for TC survivors appears to be affected by the presence of long-term treatment-related AEs.18 The relative risk of developing cardiovascular disease increases after treatment with chemotherapy. Raynaud phenomenon resulting from bleomycin-induced vascular damage developed within 4 to 12 months after chemotherapy for 18.7% to 39% of TC survivors.14,19 Bleomycin may also cause pulmonary toxicity. Pulmonary surgery, tobacco use of ≥ 20 pack-years, and a cumulative cisplatin dose of > 850 mg are risk factors for late  bleomycin-associated pulmonary toxicity.14

Other late-developing toxicities resulting from cisplatin treatment include ototoxicity, neurotoxicity, nephrotoxicity, chronic fatigue, and hypogonadism.14,19 Nearly 1 in 5 North American survivors treated with cisplatin reported severe-to-profound hearing loss within a median of 4.3 years. The extent of hearing loss has been directly associated with the increase in cumulative cisplatin dose. Peripheral neurotoxicity after cisplatin-based chemotherapy is reported to be as high as 40%.14 Chronic cancer-related fatigue can range from 15% to 27%, and has been associated with peripheral neuropathy, low testosterone levels, low physical activity, anxiety, and depression. Post-treatment hypogonadism ranges from 11% to 16%.14,17,20,21

Psychosocial issues are also of concern. Mild-to-moderate psychological distress with diagnosis and survivorship has been reported.17 Anxiety and depression are higher in TC survivors than in the general population. Variables associated with clinically significant anxiety include younger age and shorter time from diagnosis; whereas feeling helpless/hopeless, having less social support, having a higher number of physical symptoms, and having children are factors associated with higher levels of depression. A moderate-to-high level of fear of recurrence has also been reported.17

Recent Clinical Trials in Stage II Disease

Stage II disease has been the focus of current research to reduce treatment-related toxicities and limit longer-term complications. While few phase 3 clinical trials are ongoing (see Table), the results of several phase 2 trials have been reported recently.22-24

Table. Ongoing Interventional Phase 3 Clinical Trials Involving Testicular Cancer

PRIMETEST was a single-arm, single-center, phase 2 study examining the efficacy and surgical safety of primary RPLND for stage II disease.22 Participants underwent either open or robot-assisted unilateral RPLND for stage IIA or B seminoma. No adjuvant treatment was permitted. Of the 33 participants, 9 presented initially with clinical stage II disease (27%) and 24 (73%) had recurrence during active surveillance. Five of the 24 had 1 cycle of carboplatin prior to progressing to stage II. With a median follow-up of 32 months, the study did not meet its primary endpoint of PFS at 36 months. After 32 months, 10 recurrences (30%) were detected, yielding a PFS rate of 70%. All 10 patients with recurrence received chemotherapy and were alive without evidence of disease at the time of publication. This study demonstrates that RPLND may be appropriate for select patients; however, criteria for selecting patients to receive only RPLND need to be clearly defined.22

The SEMS (surgery in early metastatic seminoma) trial was a single-arm, international, phase 2 study of RPLND as first-line treatment for early metastatic seminoma with isolated retroperitoneal lymphadenopathy between 1 and 3 cm (stage II).23 With a median follow-up of 24 months, OS was 100% and 2-year recurrence-free survival was 87%. Recurrence rate was 18% (10 recurrences) with a median time to recurrence of 8 months. Short-term complications occurred in 7 patients (13%), and no patients reported long-term complications. The authors suggested that RPLND is a therapeutic option for first-line treatment in early metastatic seminoma.23

SAKK 01/10 was a single-arm, international, phase 2 study examining the de-escalation of treatment to potentially avoid toxic effects for patients with either stage IIA or stage IIB seminoma.24 Treatment included carboplatin (area under the curve [AUC] 7 mg/mL/min) followed 3 weeks later with involvednode radiotherapy (30 Gy in 15 fractions for stage IIA and 36 Gy in 18 fractions for stage IIB). The study did not meet its primary endpoint of PFS of 95% at 3 years. Grade ≥ 3 treatment-related AEs (TRAEs) included neutropenia (4%), thrombocytopenia (3%), and vomiting (1%). No treatment-related deaths and no late TRAEs were reported. One case of transient creatinine increase was reported as a serious AE, and second primary tumors were reported in 4 participants. Although the primary endpoint was not met, long-term AEs continue to be recorded for potentially up to 20 years. The favorable efficacy and toxicity profile observed in the deescalation combination treatment warrants further study.24

Emerging Trends and Future Directions for TC Treatment

Although the outlook for most newly diagnosed patients with TC is promising, especially for those diagnosed with early-stage disease and good prognosis advanced disease, treatment challenges remain. Between 10% and 20% of patients will have a relapse of TC after initially achieving a complete remission. Most patients will have a relapse within 2 years of initial treatment, but a small subgroup will have a relapse more than 5 years after therapy. Most recurrences occur in the retroperitoneum and lungs and require definitive therapy using chemotherapy and surgical resection.21

Patients with platinum-refractory disease may still achiev long-term remission with salvage therapy of surgery, conventional-dose chemotherapy, or high-dose chemotherapy with autologous stem cell transplantation; however, these treatments will fail for some patients, resulting in poor prognosis. Targeted therapy for TC has not produced meaningful benefits for this population with refractory disease, and the optimal treatment for this group of patients with TC remains to be determined.21

Although current guidelines recommend determining the levels of AFP, hCG, and LDH for clinical staging, treatment monitoring, and follow-up, limitations exist with their usage.The assays for these markers have low sensitivity and lack specificity; about half of all GCTs express only 1 of the 3 biomarkers, and seminomas lack AFP expression.7,25,26 Further research is needed on LDH. An emerging group of patients with LDH below 2.5× ULN may be candidates for de-escalatio strategies to reduce treatment burden, while inferior outcomes remain for patients with either good prognosis seminoma and elevated LDH, or intermediate prognosis seminoma.7

Other biomarkers, such as miRNA371a-3p and PD-L1, are being investigated; miRNA371a-3p has been shown to have prognostic significance. The results of this assay can be informative for both seminomas and NSGCTs.26 However, the protocol for quantification and implementation still needs to be determined.27

Click to read more from 2023 Rare Diseases Report: Cancers

References
  1. Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA Cancer J Clin. 2023;73(1):17-48. doi:10.3322/caac.21763
  2. Cheng L, Albers P, Berney DM, et al. Testicular cancer. Nat Rev Dis Primers. 2018;4(1):29. doi:10.1038/s41572-018-0029-03
  3. Chovanec M, Cheng L. Advances in diagnosis and treatment of testicular cancer. BMJ. 2022;379:e070499. doi:10.1136/bmj-2022-070499
  4. Gaddam SJ, Chesnut GT. Testicle cancer. StatPearls [Internet]. Treasure Island, FL: StatPearls Publishing; 2022. Updated October 16, 2022. Accessed March 13, 2023. https://www.ncbi.nlm.nih.gov/books/NBK563159/
  5. Yang H, Obiora D, Tomaszewski JJ. Outcomes and expanding indications for robotic retroperitoneal lymph node dissection for testicular cancer. Transl Androl Urol. 2021;10(5):2188-2194. doi:10.21037/tau.2020.03.14
  6. International Germ Cell Cancer Collaborative Group. International germ cell consensus classification: a prognostic factor-based staging system for metastatic germ cell cancers. J Clin Oncol. 1997;15(2):594-603. doi:10.1200/JCO.1997.15.2.594
  7. Beyer J, Collette L, Sauvé N, et al. Survival and new prognosticators in metastatic seminoma: results from the IGCCCG-Update Consortium. J Clin Oncol. 2021;39(14):1553-1562. doi:10.1200/JCO.20.03292
  8. Gillessen S, Sauvé N, Collette L, et al. Predicting outcomes in men with metastatic nonseminomatous germ cell tumors (NSGCT): results from the IGCCCG Update Consortium. J Clin Oncol. 2021;39(14):1563-1574. doi:10.1200/JCO.20.03296
  9. Gilligan T, Lin DW, Aggarwal R, et al. Testicular cancer, version 2.2020, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2019;17(12):1529-1554. doi:10.6004/jnccn.2019.0058
  10. Heinzelbecker J, Schmidt S, Lackner J, et al. Therapy of clinical stage IIa and IIb seminoma: a systematic review. World J Urol. 2022;40(12):2829-2841. doi:10.1007/s00345-021-03873-5
  11. Oldenburg J, Berney DM, Bokemeyer C, et al. Testicular seminoma and nonseminoma: ESMA-EURACAN Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol. 2022;33(4):362-375. doi:10.1016/j.annonc.2022.01.002
  12.  Wymer KM, Pearce SM, Harris KT, Pierorazio PM, Daneshmand S, Eggener SE. Adherence to National Comprehensive Cancer Network® guidelines for testicular cancer. J Urol. 2017;197(3 pt 1):684-689. doi:10.1016/j.juro.2016.09.073
  13. Saoud RM, Andolfi C, Aizen J, et al. Impact of non-guideline-directed care on quality of life in testicular cancer survivors. Eur Urol Focus. 2021;7(5):1137-1142. doi:10.1016/j.euf.2020.10.005
  14. Fung C, Dinh PC, Fossa SD, Travis LB. Testicular cancer survivorship. J Natl Compr Canc Netw. 2019;17(12):1557-1568. doi:10.6004/jnccn.2019.7369
  15. Guo CC, Czerniak B. Somatic-type malignancies in testicular germ cell tumors. Hum Pathol. 2022;127:123-135.
  16. Sprauten M, Darrah TH, Peterson DR, et al. Impact of long-term serum platinum concentrations on neuro- and ototoxicity in cisplatin-treated survivors of testicular cancer. J Clin Oncol. 2012;30(3):300-307. doi:10.1200/JCO.2011.37.4025
  17. Shrem NS, Wood L, Hamilton RJ, et al. Testicular cancer survivorship: long-term toxicity and management. Can Urol Assoc J. 2022;16(8):257-272. doi:10.5489/cuaj.8009
  18. Hellesnes R, Myklebust TA, Fosså SD, et al. Testicular cancer in the cisplatin era: causes of death and mortality rates in a population-based cohort. J Clin Oncol. 2021;39(32):3561-3573. doi:10.1200/JCO.21.00637
  19. Mercieca-Bebber R, Naher SK, Rincones O, Smith AB, Stockler MR. Patient-reported outcomes associated with treatments for testicular cancer: a systematic review. Patient Relat Outcome Meas. 2021;12:129-171. doi:10.2147/PROM.S242754
  20. Sprauten M, Haugnes HS, Brydøy M, et al. Chronic fatigue in 812 testicular cancer survivors during long-term follow-up: increasing prevalence and risk factors. Ann Oncol. 2015;26(10):2133-2140. doi:10.1093/annonc/mdv328
  21. King J, Adra N, Einhorn LH. Testicular cancer: biology to bedside. Cancer Res. 2021;81(21):5369-5376. doi:10.1158/0008-5472.CAN-21-1452
  22. Hiester A, Che Y, Lusch A, et al. Phase 2 single-arm trial of primary retroperitoneal lymph node dissection in patients with seminomatous testicular germ cell tumors with clinical stage IIA/B (PRIMETEST). Eur Urol. 2022;S0302-2838(22)02775-0. doi:10.1016/j.eururo.2022.10.021
  23. Daneshmand S, Cary C, Masterson TA, et al. SEMS trial: result of a prospective, multi-institutional phase II clinical trial of surgery in early metastatic seminoma. J Clin Oncol. 2021;39(6 suppl):Abstract 375. doi:10.1200JCO.2021.39.6_suppl.375
  24. Papachristofilou A, Bedke J, Hayoz S, et al. Single-dose carboplatin followed by involved-node radiotherapy for stage IIA and stage IIB seminoma (SAKK 01/10): a single-arm, multicentre, phase 2 trial. Lancet Oncol. 2022;23(11):1441-1450. doi:10.1016/S1470-2045(22)00564-2
  25. Dieckmann KP, Richter-Simonsen H, Kulejewski M, et al. Testicular germ-cell tumours: a descriptive analysis of clinical characteristics at first presentation. Urol Int. 2018;100(4):409-419. doi:10.1159/000488284
  26. Murray MJ, Huddart RA, Coleman N. The present and future of serum diagnostic tests for testicular germ cell tumours. Nat Rev Urol. 2016;13(12):715-725. doi:10.1038/nrurol.2016.170
Author and Disclosure Information

Liang Cheng, MD
Professor and Vice Chair for Translational Research
Director of Anatomic and Molecular Pathology
Department of Pathology and Laboratory Medicine
Brown University Warren Alpert Medical School, Lifespan Academic Medical Center
Legorreta Cancer Center at Brown University
Providence, RI

Liang Cheng, MD, has disclosed no relevant financial relationships.

Publications
Topics
Author and Disclosure Information

Liang Cheng, MD
Professor and Vice Chair for Translational Research
Director of Anatomic and Molecular Pathology
Department of Pathology and Laboratory Medicine
Brown University Warren Alpert Medical School, Lifespan Academic Medical Center
Legorreta Cancer Center at Brown University
Providence, RI

Liang Cheng, MD, has disclosed no relevant financial relationships.

Author and Disclosure Information

Liang Cheng, MD
Professor and Vice Chair for Translational Research
Director of Anatomic and Molecular Pathology
Department of Pathology and Laboratory Medicine
Brown University Warren Alpert Medical School, Lifespan Academic Medical Center
Legorreta Cancer Center at Brown University
Providence, RI

Liang Cheng, MD, has disclosed no relevant financial relationships.

Liang Cheng, MD
Approximately 1% of adult neoplasms and 5% of all urologic cancers are testicular cancer (TC).1 In the United States, 9190 new cases have been estimated for 2023.1 Testicular germ cell tumors (GCTs) comprise 90% to 95% of all TCs and are grouped into seminomas, nonseminomatous GCTs (NSGCTs), and mixed histology GCTs.1 NSGCTs tend to be more aggressive and are more common in younger men (15-40 years old), whereas seminomas are slower growing and generally develop later in a patient’s life.2,3 


Mortality from TC has been decreasing since the 1970s due to cisplatin-based chemotherapy regimens2,3; TC is among the most curable of solid neoplasms, with a 5-year relative survival rate of 95%.2-4 Thus, the focus of research has shifted from optimizing treatments for improved survival to decreasing treatment-related, long-term adverse events (AEs).5

New Modifications in Risk Assessment and Prognostication

The widely accepted risk stratification model in use today was first developed in 1997 by the International Germ Cell Cancer Collaborative Group (IGCCCG) after studying data on patients with seminoma and NSGCTs.6 The original classification categorized metastatic NSGCTs as having good, intermediate, or poor prognosis based on levels of alpha-fetoprotein (AFP), human chorionic gonadotropin (hCG), lactate dehydrogenase (LDH), and the presence of nonpulmonary visceral metastases (NPVM). Primary mediastinal NSGCTs were classified as having poor prognosis regardless of the other factors.Metastatic seminoma GCTs were categorized as having good or intermediate prognosis based on the occurrence of brain, liver, or bone metastasis.7

Using contemporary data from more than 12,000 patients with metastatic GCTs who received either cisplatin or etoposide, the IGCCCG model was updated in 2021. For seminoma GCTs, 5-year progression-free survival (PFS) and 5-year overall survival (OS) were extended for both good and intermediate prognostic groups.7 LDH remained the most significant prognostic factor for determining good prognosis however, patients with LDH above 2.5× upper limit of normal (ULN) before chemotherapy had worse survival probabilities than patients with LDH at 2.5× ULN or lower. The survival probabilities for patients with otherwise good prognosis with LDH of more than 2.5× ULN were like those for patients with intermediate prognosis.7 Thus, using LDH of more than 2.5× ULN has revealed a subgroup with significantly worse outcomes within the “good” prognostic group.7,8

For NSGCTs, 5-year PFS rates did not differ from the original IGCCCG for good and intermediate prognostic groups; however, the 2021 update revealed an improved PFS for the poor prognostic group. The 2021 update also demonstrated that 5-year OS rates improved for each group, and further confirmed that the 2 most important prognostic factors for NSGCT were the presence of NPVM and the presence of a mediastinal primary tumor. The update added 2 new adverse prognostic variables: age and metastases. Risk of progression increases 25% with every decade-of-life increase, and 66% with the presence of lung metastases. The LDH groups were reduced to a single cutoff at 2.5× ULN for NSGCTs.8

Primary and Subsequent Treatments for TC

Guideline-directed first-line and subsequent treatments for seminomas and NSGCTs have been developed by several organizations, including the National Comprehensive Cancer Network, IGCCCG, and the American Urological Association (see Figure 1 and 2). An analysis of the most used treatments was performed using the National Cancer Database.2 Most patients underwent orchiectomy without chemotherapy or radiation for both stage I seminomas (78%) and NSGCTs (57%). For stage II and III seminomas, most patients underwent surgery with chemotherapy (66% and 68%, respectively). Nearly half of patients with stage II NSGCTs were treated with surgery and chemotherapy (49%), and a third were treated with retroperitoneal lymph node dissection (RPLND) in addition to surgery and chemotherapy. Surgery with chemotherapy was used for 55% of stage III NSGCTs; other treatments included surgery combined with chemotherapy and RPLND (19%), and chemotherapy with or without radiation (20%).2 However, nearly 30% of patients with TC do not receive guideline-directed therapy, including inappropriate imaging and overtreatment; and nonguideline–directed therapy has been independently associated with risk of relapse.12,13

Figure 1. First-line and Subsequent Treatment of Seminomas

Figure 2. First-line and Subsequent Treatment of Non-Seminomas

TC Survivorship

The trend of improved OS after treatment for metastatic GCTs highlights a need to focus on survivorship. The 10-year survival rate for TC post-treatment is 95%.14 Latest estimates suggest there are more than 300,000 TC survivors in the United States,accounting for approximately 4% of all US male cancer survivors.14 With longer-term survival, however, comes the risk for long-term complications from cancer treatments. For example, circulating platinum has been detected in the plasma of men up to 28 years after undergoing cisplatin-based chemotherapy for TC.15 Increasing levels of residual serum platinum have also been shown to correlate with severity of neurotoxicity between 5 and 20 years after treatment.16

A significant concern with cancer treatment is the development of second malignant neoplasms (SMNs).14,17 The relative risk of the development of SMNs depends on whetherradiation therapy or chemotherapy, or both, was used as the primary treatment. Patients who received either radiation therapy or chemotherapy are at increased risk for leukemia and solid cancers, including gastrointestinal cancers. For patients treated with cisplatin, a significant dose-response relationship between cumulative dose and leukemic risk has been reported.14

Other concerns are increased non-TC mortality and SMN mortality. Hellesnes et al examined cause-specific, non-TC mortality using a population-based cohort in Norway.18 They determined that the overall 25-year, non-TC mortality risk was 13.7% (95% CI, 12.5-14.9) for patients who previously had TC vs 11.3% for patients who never had TC. The highest mortality rates were reported for patients who had radiation (19%) or platinum-based chemotherapy plus radiation (18.4%); the lowest mortality rate was reported for patients who had received platinum-based chemotherapy only (9.5%). Patients with the highest non-TC mortality risk were fewer than 20 years post-cancer diagnosis. Non-TC mortality excess ranged from 23% to 40% for patients with a prior TC diagnosis, and a significant 1.43- to 3.24-fold increase in SMN mortality emerged after treatment with platinum-based chemotherapy or radiation therapy, or both.19 Awareness of the increased premature mortality risk is crucial for both TC survivors and their care providers.18

Quality of life for TC survivors appears to be affected by the presence of long-term treatment-related AEs.18 The relative risk of developing cardiovascular disease increases after treatment with chemotherapy. Raynaud phenomenon resulting from bleomycin-induced vascular damage developed within 4 to 12 months after chemotherapy for 18.7% to 39% of TC survivors.14,19 Bleomycin may also cause pulmonary toxicity. Pulmonary surgery, tobacco use of ≥ 20 pack-years, and a cumulative cisplatin dose of > 850 mg are risk factors for late  bleomycin-associated pulmonary toxicity.14

Other late-developing toxicities resulting from cisplatin treatment include ototoxicity, neurotoxicity, nephrotoxicity, chronic fatigue, and hypogonadism.14,19 Nearly 1 in 5 North American survivors treated with cisplatin reported severe-to-profound hearing loss within a median of 4.3 years. The extent of hearing loss has been directly associated with the increase in cumulative cisplatin dose. Peripheral neurotoxicity after cisplatin-based chemotherapy is reported to be as high as 40%.14 Chronic cancer-related fatigue can range from 15% to 27%, and has been associated with peripheral neuropathy, low testosterone levels, low physical activity, anxiety, and depression. Post-treatment hypogonadism ranges from 11% to 16%.14,17,20,21

Psychosocial issues are also of concern. Mild-to-moderate psychological distress with diagnosis and survivorship has been reported.17 Anxiety and depression are higher in TC survivors than in the general population. Variables associated with clinically significant anxiety include younger age and shorter time from diagnosis; whereas feeling helpless/hopeless, having less social support, having a higher number of physical symptoms, and having children are factors associated with higher levels of depression. A moderate-to-high level of fear of recurrence has also been reported.17

Recent Clinical Trials in Stage II Disease

Stage II disease has been the focus of current research to reduce treatment-related toxicities and limit longer-term complications. While few phase 3 clinical trials are ongoing (see Table), the results of several phase 2 trials have been reported recently.22-24

Table. Ongoing Interventional Phase 3 Clinical Trials Involving Testicular Cancer

PRIMETEST was a single-arm, single-center, phase 2 study examining the efficacy and surgical safety of primary RPLND for stage II disease.22 Participants underwent either open or robot-assisted unilateral RPLND for stage IIA or B seminoma. No adjuvant treatment was permitted. Of the 33 participants, 9 presented initially with clinical stage II disease (27%) and 24 (73%) had recurrence during active surveillance. Five of the 24 had 1 cycle of carboplatin prior to progressing to stage II. With a median follow-up of 32 months, the study did not meet its primary endpoint of PFS at 36 months. After 32 months, 10 recurrences (30%) were detected, yielding a PFS rate of 70%. All 10 patients with recurrence received chemotherapy and were alive without evidence of disease at the time of publication. This study demonstrates that RPLND may be appropriate for select patients; however, criteria for selecting patients to receive only RPLND need to be clearly defined.22

The SEMS (surgery in early metastatic seminoma) trial was a single-arm, international, phase 2 study of RPLND as first-line treatment for early metastatic seminoma with isolated retroperitoneal lymphadenopathy between 1 and 3 cm (stage II).23 With a median follow-up of 24 months, OS was 100% and 2-year recurrence-free survival was 87%. Recurrence rate was 18% (10 recurrences) with a median time to recurrence of 8 months. Short-term complications occurred in 7 patients (13%), and no patients reported long-term complications. The authors suggested that RPLND is a therapeutic option for first-line treatment in early metastatic seminoma.23

SAKK 01/10 was a single-arm, international, phase 2 study examining the de-escalation of treatment to potentially avoid toxic effects for patients with either stage IIA or stage IIB seminoma.24 Treatment included carboplatin (area under the curve [AUC] 7 mg/mL/min) followed 3 weeks later with involvednode radiotherapy (30 Gy in 15 fractions for stage IIA and 36 Gy in 18 fractions for stage IIB). The study did not meet its primary endpoint of PFS of 95% at 3 years. Grade ≥ 3 treatment-related AEs (TRAEs) included neutropenia (4%), thrombocytopenia (3%), and vomiting (1%). No treatment-related deaths and no late TRAEs were reported. One case of transient creatinine increase was reported as a serious AE, and second primary tumors were reported in 4 participants. Although the primary endpoint was not met, long-term AEs continue to be recorded for potentially up to 20 years. The favorable efficacy and toxicity profile observed in the deescalation combination treatment warrants further study.24

Emerging Trends and Future Directions for TC Treatment

Although the outlook for most newly diagnosed patients with TC is promising, especially for those diagnosed with early-stage disease and good prognosis advanced disease, treatment challenges remain. Between 10% and 20% of patients will have a relapse of TC after initially achieving a complete remission. Most patients will have a relapse within 2 years of initial treatment, but a small subgroup will have a relapse more than 5 years after therapy. Most recurrences occur in the retroperitoneum and lungs and require definitive therapy using chemotherapy and surgical resection.21

Patients with platinum-refractory disease may still achiev long-term remission with salvage therapy of surgery, conventional-dose chemotherapy, or high-dose chemotherapy with autologous stem cell transplantation; however, these treatments will fail for some patients, resulting in poor prognosis. Targeted therapy for TC has not produced meaningful benefits for this population with refractory disease, and the optimal treatment for this group of patients with TC remains to be determined.21

Although current guidelines recommend determining the levels of AFP, hCG, and LDH for clinical staging, treatment monitoring, and follow-up, limitations exist with their usage.The assays for these markers have low sensitivity and lack specificity; about half of all GCTs express only 1 of the 3 biomarkers, and seminomas lack AFP expression.7,25,26 Further research is needed on LDH. An emerging group of patients with LDH below 2.5× ULN may be candidates for de-escalatio strategies to reduce treatment burden, while inferior outcomes remain for patients with either good prognosis seminoma and elevated LDH, or intermediate prognosis seminoma.7

Other biomarkers, such as miRNA371a-3p and PD-L1, are being investigated; miRNA371a-3p has been shown to have prognostic significance. The results of this assay can be informative for both seminomas and NSGCTs.26 However, the protocol for quantification and implementation still needs to be determined.27

Click to read more from 2023 Rare Diseases Report: Cancers

Liang Cheng, MD
Approximately 1% of adult neoplasms and 5% of all urologic cancers are testicular cancer (TC).1 In the United States, 9190 new cases have been estimated for 2023.1 Testicular germ cell tumors (GCTs) comprise 90% to 95% of all TCs and are grouped into seminomas, nonseminomatous GCTs (NSGCTs), and mixed histology GCTs.1 NSGCTs tend to be more aggressive and are more common in younger men (15-40 years old), whereas seminomas are slower growing and generally develop later in a patient’s life.2,3 


Mortality from TC has been decreasing since the 1970s due to cisplatin-based chemotherapy regimens2,3; TC is among the most curable of solid neoplasms, with a 5-year relative survival rate of 95%.2-4 Thus, the focus of research has shifted from optimizing treatments for improved survival to decreasing treatment-related, long-term adverse events (AEs).5

New Modifications in Risk Assessment and Prognostication

The widely accepted risk stratification model in use today was first developed in 1997 by the International Germ Cell Cancer Collaborative Group (IGCCCG) after studying data on patients with seminoma and NSGCTs.6 The original classification categorized metastatic NSGCTs as having good, intermediate, or poor prognosis based on levels of alpha-fetoprotein (AFP), human chorionic gonadotropin (hCG), lactate dehydrogenase (LDH), and the presence of nonpulmonary visceral metastases (NPVM). Primary mediastinal NSGCTs were classified as having poor prognosis regardless of the other factors.Metastatic seminoma GCTs were categorized as having good or intermediate prognosis based on the occurrence of brain, liver, or bone metastasis.7

Using contemporary data from more than 12,000 patients with metastatic GCTs who received either cisplatin or etoposide, the IGCCCG model was updated in 2021. For seminoma GCTs, 5-year progression-free survival (PFS) and 5-year overall survival (OS) were extended for both good and intermediate prognostic groups.7 LDH remained the most significant prognostic factor for determining good prognosis however, patients with LDH above 2.5× upper limit of normal (ULN) before chemotherapy had worse survival probabilities than patients with LDH at 2.5× ULN or lower. The survival probabilities for patients with otherwise good prognosis with LDH of more than 2.5× ULN were like those for patients with intermediate prognosis.7 Thus, using LDH of more than 2.5× ULN has revealed a subgroup with significantly worse outcomes within the “good” prognostic group.7,8

For NSGCTs, 5-year PFS rates did not differ from the original IGCCCG for good and intermediate prognostic groups; however, the 2021 update revealed an improved PFS for the poor prognostic group. The 2021 update also demonstrated that 5-year OS rates improved for each group, and further confirmed that the 2 most important prognostic factors for NSGCT were the presence of NPVM and the presence of a mediastinal primary tumor. The update added 2 new adverse prognostic variables: age and metastases. Risk of progression increases 25% with every decade-of-life increase, and 66% with the presence of lung metastases. The LDH groups were reduced to a single cutoff at 2.5× ULN for NSGCTs.8

Primary and Subsequent Treatments for TC

Guideline-directed first-line and subsequent treatments for seminomas and NSGCTs have been developed by several organizations, including the National Comprehensive Cancer Network, IGCCCG, and the American Urological Association (see Figure 1 and 2). An analysis of the most used treatments was performed using the National Cancer Database.2 Most patients underwent orchiectomy without chemotherapy or radiation for both stage I seminomas (78%) and NSGCTs (57%). For stage II and III seminomas, most patients underwent surgery with chemotherapy (66% and 68%, respectively). Nearly half of patients with stage II NSGCTs were treated with surgery and chemotherapy (49%), and a third were treated with retroperitoneal lymph node dissection (RPLND) in addition to surgery and chemotherapy. Surgery with chemotherapy was used for 55% of stage III NSGCTs; other treatments included surgery combined with chemotherapy and RPLND (19%), and chemotherapy with or without radiation (20%).2 However, nearly 30% of patients with TC do not receive guideline-directed therapy, including inappropriate imaging and overtreatment; and nonguideline–directed therapy has been independently associated with risk of relapse.12,13

Figure 1. First-line and Subsequent Treatment of Seminomas

Figure 2. First-line and Subsequent Treatment of Non-Seminomas

TC Survivorship

The trend of improved OS after treatment for metastatic GCTs highlights a need to focus on survivorship. The 10-year survival rate for TC post-treatment is 95%.14 Latest estimates suggest there are more than 300,000 TC survivors in the United States,accounting for approximately 4% of all US male cancer survivors.14 With longer-term survival, however, comes the risk for long-term complications from cancer treatments. For example, circulating platinum has been detected in the plasma of men up to 28 years after undergoing cisplatin-based chemotherapy for TC.15 Increasing levels of residual serum platinum have also been shown to correlate with severity of neurotoxicity between 5 and 20 years after treatment.16

A significant concern with cancer treatment is the development of second malignant neoplasms (SMNs).14,17 The relative risk of the development of SMNs depends on whetherradiation therapy or chemotherapy, or both, was used as the primary treatment. Patients who received either radiation therapy or chemotherapy are at increased risk for leukemia and solid cancers, including gastrointestinal cancers. For patients treated with cisplatin, a significant dose-response relationship between cumulative dose and leukemic risk has been reported.14

Other concerns are increased non-TC mortality and SMN mortality. Hellesnes et al examined cause-specific, non-TC mortality using a population-based cohort in Norway.18 They determined that the overall 25-year, non-TC mortality risk was 13.7% (95% CI, 12.5-14.9) for patients who previously had TC vs 11.3% for patients who never had TC. The highest mortality rates were reported for patients who had radiation (19%) or platinum-based chemotherapy plus radiation (18.4%); the lowest mortality rate was reported for patients who had received platinum-based chemotherapy only (9.5%). Patients with the highest non-TC mortality risk were fewer than 20 years post-cancer diagnosis. Non-TC mortality excess ranged from 23% to 40% for patients with a prior TC diagnosis, and a significant 1.43- to 3.24-fold increase in SMN mortality emerged after treatment with platinum-based chemotherapy or radiation therapy, or both.19 Awareness of the increased premature mortality risk is crucial for both TC survivors and their care providers.18

Quality of life for TC survivors appears to be affected by the presence of long-term treatment-related AEs.18 The relative risk of developing cardiovascular disease increases after treatment with chemotherapy. Raynaud phenomenon resulting from bleomycin-induced vascular damage developed within 4 to 12 months after chemotherapy for 18.7% to 39% of TC survivors.14,19 Bleomycin may also cause pulmonary toxicity. Pulmonary surgery, tobacco use of ≥ 20 pack-years, and a cumulative cisplatin dose of > 850 mg are risk factors for late  bleomycin-associated pulmonary toxicity.14

Other late-developing toxicities resulting from cisplatin treatment include ototoxicity, neurotoxicity, nephrotoxicity, chronic fatigue, and hypogonadism.14,19 Nearly 1 in 5 North American survivors treated with cisplatin reported severe-to-profound hearing loss within a median of 4.3 years. The extent of hearing loss has been directly associated with the increase in cumulative cisplatin dose. Peripheral neurotoxicity after cisplatin-based chemotherapy is reported to be as high as 40%.14 Chronic cancer-related fatigue can range from 15% to 27%, and has been associated with peripheral neuropathy, low testosterone levels, low physical activity, anxiety, and depression. Post-treatment hypogonadism ranges from 11% to 16%.14,17,20,21

Psychosocial issues are also of concern. Mild-to-moderate psychological distress with diagnosis and survivorship has been reported.17 Anxiety and depression are higher in TC survivors than in the general population. Variables associated with clinically significant anxiety include younger age and shorter time from diagnosis; whereas feeling helpless/hopeless, having less social support, having a higher number of physical symptoms, and having children are factors associated with higher levels of depression. A moderate-to-high level of fear of recurrence has also been reported.17

Recent Clinical Trials in Stage II Disease

Stage II disease has been the focus of current research to reduce treatment-related toxicities and limit longer-term complications. While few phase 3 clinical trials are ongoing (see Table), the results of several phase 2 trials have been reported recently.22-24

Table. Ongoing Interventional Phase 3 Clinical Trials Involving Testicular Cancer

PRIMETEST was a single-arm, single-center, phase 2 study examining the efficacy and surgical safety of primary RPLND for stage II disease.22 Participants underwent either open or robot-assisted unilateral RPLND for stage IIA or B seminoma. No adjuvant treatment was permitted. Of the 33 participants, 9 presented initially with clinical stage II disease (27%) and 24 (73%) had recurrence during active surveillance. Five of the 24 had 1 cycle of carboplatin prior to progressing to stage II. With a median follow-up of 32 months, the study did not meet its primary endpoint of PFS at 36 months. After 32 months, 10 recurrences (30%) were detected, yielding a PFS rate of 70%. All 10 patients with recurrence received chemotherapy and were alive without evidence of disease at the time of publication. This study demonstrates that RPLND may be appropriate for select patients; however, criteria for selecting patients to receive only RPLND need to be clearly defined.22

The SEMS (surgery in early metastatic seminoma) trial was a single-arm, international, phase 2 study of RPLND as first-line treatment for early metastatic seminoma with isolated retroperitoneal lymphadenopathy between 1 and 3 cm (stage II).23 With a median follow-up of 24 months, OS was 100% and 2-year recurrence-free survival was 87%. Recurrence rate was 18% (10 recurrences) with a median time to recurrence of 8 months. Short-term complications occurred in 7 patients (13%), and no patients reported long-term complications. The authors suggested that RPLND is a therapeutic option for first-line treatment in early metastatic seminoma.23

SAKK 01/10 was a single-arm, international, phase 2 study examining the de-escalation of treatment to potentially avoid toxic effects for patients with either stage IIA or stage IIB seminoma.24 Treatment included carboplatin (area under the curve [AUC] 7 mg/mL/min) followed 3 weeks later with involvednode radiotherapy (30 Gy in 15 fractions for stage IIA and 36 Gy in 18 fractions for stage IIB). The study did not meet its primary endpoint of PFS of 95% at 3 years. Grade ≥ 3 treatment-related AEs (TRAEs) included neutropenia (4%), thrombocytopenia (3%), and vomiting (1%). No treatment-related deaths and no late TRAEs were reported. One case of transient creatinine increase was reported as a serious AE, and second primary tumors were reported in 4 participants. Although the primary endpoint was not met, long-term AEs continue to be recorded for potentially up to 20 years. The favorable efficacy and toxicity profile observed in the deescalation combination treatment warrants further study.24

Emerging Trends and Future Directions for TC Treatment

Although the outlook for most newly diagnosed patients with TC is promising, especially for those diagnosed with early-stage disease and good prognosis advanced disease, treatment challenges remain. Between 10% and 20% of patients will have a relapse of TC after initially achieving a complete remission. Most patients will have a relapse within 2 years of initial treatment, but a small subgroup will have a relapse more than 5 years after therapy. Most recurrences occur in the retroperitoneum and lungs and require definitive therapy using chemotherapy and surgical resection.21

Patients with platinum-refractory disease may still achiev long-term remission with salvage therapy of surgery, conventional-dose chemotherapy, or high-dose chemotherapy with autologous stem cell transplantation; however, these treatments will fail for some patients, resulting in poor prognosis. Targeted therapy for TC has not produced meaningful benefits for this population with refractory disease, and the optimal treatment for this group of patients with TC remains to be determined.21

Although current guidelines recommend determining the levels of AFP, hCG, and LDH for clinical staging, treatment monitoring, and follow-up, limitations exist with their usage.The assays for these markers have low sensitivity and lack specificity; about half of all GCTs express only 1 of the 3 biomarkers, and seminomas lack AFP expression.7,25,26 Further research is needed on LDH. An emerging group of patients with LDH below 2.5× ULN may be candidates for de-escalatio strategies to reduce treatment burden, while inferior outcomes remain for patients with either good prognosis seminoma and elevated LDH, or intermediate prognosis seminoma.7

Other biomarkers, such as miRNA371a-3p and PD-L1, are being investigated; miRNA371a-3p has been shown to have prognostic significance. The results of this assay can be informative for both seminomas and NSGCTs.26 However, the protocol for quantification and implementation still needs to be determined.27

Click to read more from 2023 Rare Diseases Report: Cancers

References
  1. Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA Cancer J Clin. 2023;73(1):17-48. doi:10.3322/caac.21763
  2. Cheng L, Albers P, Berney DM, et al. Testicular cancer. Nat Rev Dis Primers. 2018;4(1):29. doi:10.1038/s41572-018-0029-03
  3. Chovanec M, Cheng L. Advances in diagnosis and treatment of testicular cancer. BMJ. 2022;379:e070499. doi:10.1136/bmj-2022-070499
  4. Gaddam SJ, Chesnut GT. Testicle cancer. StatPearls [Internet]. Treasure Island, FL: StatPearls Publishing; 2022. Updated October 16, 2022. Accessed March 13, 2023. https://www.ncbi.nlm.nih.gov/books/NBK563159/
  5. Yang H, Obiora D, Tomaszewski JJ. Outcomes and expanding indications for robotic retroperitoneal lymph node dissection for testicular cancer. Transl Androl Urol. 2021;10(5):2188-2194. doi:10.21037/tau.2020.03.14
  6. International Germ Cell Cancer Collaborative Group. International germ cell consensus classification: a prognostic factor-based staging system for metastatic germ cell cancers. J Clin Oncol. 1997;15(2):594-603. doi:10.1200/JCO.1997.15.2.594
  7. Beyer J, Collette L, Sauvé N, et al. Survival and new prognosticators in metastatic seminoma: results from the IGCCCG-Update Consortium. J Clin Oncol. 2021;39(14):1553-1562. doi:10.1200/JCO.20.03292
  8. Gillessen S, Sauvé N, Collette L, et al. Predicting outcomes in men with metastatic nonseminomatous germ cell tumors (NSGCT): results from the IGCCCG Update Consortium. J Clin Oncol. 2021;39(14):1563-1574. doi:10.1200/JCO.20.03296
  9. Gilligan T, Lin DW, Aggarwal R, et al. Testicular cancer, version 2.2020, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2019;17(12):1529-1554. doi:10.6004/jnccn.2019.0058
  10. Heinzelbecker J, Schmidt S, Lackner J, et al. Therapy of clinical stage IIa and IIb seminoma: a systematic review. World J Urol. 2022;40(12):2829-2841. doi:10.1007/s00345-021-03873-5
  11. Oldenburg J, Berney DM, Bokemeyer C, et al. Testicular seminoma and nonseminoma: ESMA-EURACAN Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol. 2022;33(4):362-375. doi:10.1016/j.annonc.2022.01.002
  12.  Wymer KM, Pearce SM, Harris KT, Pierorazio PM, Daneshmand S, Eggener SE. Adherence to National Comprehensive Cancer Network® guidelines for testicular cancer. J Urol. 2017;197(3 pt 1):684-689. doi:10.1016/j.juro.2016.09.073
  13. Saoud RM, Andolfi C, Aizen J, et al. Impact of non-guideline-directed care on quality of life in testicular cancer survivors. Eur Urol Focus. 2021;7(5):1137-1142. doi:10.1016/j.euf.2020.10.005
  14. Fung C, Dinh PC, Fossa SD, Travis LB. Testicular cancer survivorship. J Natl Compr Canc Netw. 2019;17(12):1557-1568. doi:10.6004/jnccn.2019.7369
  15. Guo CC, Czerniak B. Somatic-type malignancies in testicular germ cell tumors. Hum Pathol. 2022;127:123-135.
  16. Sprauten M, Darrah TH, Peterson DR, et al. Impact of long-term serum platinum concentrations on neuro- and ototoxicity in cisplatin-treated survivors of testicular cancer. J Clin Oncol. 2012;30(3):300-307. doi:10.1200/JCO.2011.37.4025
  17. Shrem NS, Wood L, Hamilton RJ, et al. Testicular cancer survivorship: long-term toxicity and management. Can Urol Assoc J. 2022;16(8):257-272. doi:10.5489/cuaj.8009
  18. Hellesnes R, Myklebust TA, Fosså SD, et al. Testicular cancer in the cisplatin era: causes of death and mortality rates in a population-based cohort. J Clin Oncol. 2021;39(32):3561-3573. doi:10.1200/JCO.21.00637
  19. Mercieca-Bebber R, Naher SK, Rincones O, Smith AB, Stockler MR. Patient-reported outcomes associated with treatments for testicular cancer: a systematic review. Patient Relat Outcome Meas. 2021;12:129-171. doi:10.2147/PROM.S242754
  20. Sprauten M, Haugnes HS, Brydøy M, et al. Chronic fatigue in 812 testicular cancer survivors during long-term follow-up: increasing prevalence and risk factors. Ann Oncol. 2015;26(10):2133-2140. doi:10.1093/annonc/mdv328
  21. King J, Adra N, Einhorn LH. Testicular cancer: biology to bedside. Cancer Res. 2021;81(21):5369-5376. doi:10.1158/0008-5472.CAN-21-1452
  22. Hiester A, Che Y, Lusch A, et al. Phase 2 single-arm trial of primary retroperitoneal lymph node dissection in patients with seminomatous testicular germ cell tumors with clinical stage IIA/B (PRIMETEST). Eur Urol. 2022;S0302-2838(22)02775-0. doi:10.1016/j.eururo.2022.10.021
  23. Daneshmand S, Cary C, Masterson TA, et al. SEMS trial: result of a prospective, multi-institutional phase II clinical trial of surgery in early metastatic seminoma. J Clin Oncol. 2021;39(6 suppl):Abstract 375. doi:10.1200JCO.2021.39.6_suppl.375
  24. Papachristofilou A, Bedke J, Hayoz S, et al. Single-dose carboplatin followed by involved-node radiotherapy for stage IIA and stage IIB seminoma (SAKK 01/10): a single-arm, multicentre, phase 2 trial. Lancet Oncol. 2022;23(11):1441-1450. doi:10.1016/S1470-2045(22)00564-2
  25. Dieckmann KP, Richter-Simonsen H, Kulejewski M, et al. Testicular germ-cell tumours: a descriptive analysis of clinical characteristics at first presentation. Urol Int. 2018;100(4):409-419. doi:10.1159/000488284
  26. Murray MJ, Huddart RA, Coleman N. The present and future of serum diagnostic tests for testicular germ cell tumours. Nat Rev Urol. 2016;13(12):715-725. doi:10.1038/nrurol.2016.170
References
  1. Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA Cancer J Clin. 2023;73(1):17-48. doi:10.3322/caac.21763
  2. Cheng L, Albers P, Berney DM, et al. Testicular cancer. Nat Rev Dis Primers. 2018;4(1):29. doi:10.1038/s41572-018-0029-03
  3. Chovanec M, Cheng L. Advances in diagnosis and treatment of testicular cancer. BMJ. 2022;379:e070499. doi:10.1136/bmj-2022-070499
  4. Gaddam SJ, Chesnut GT. Testicle cancer. StatPearls [Internet]. Treasure Island, FL: StatPearls Publishing; 2022. Updated October 16, 2022. Accessed March 13, 2023. https://www.ncbi.nlm.nih.gov/books/NBK563159/
  5. Yang H, Obiora D, Tomaszewski JJ. Outcomes and expanding indications for robotic retroperitoneal lymph node dissection for testicular cancer. Transl Androl Urol. 2021;10(5):2188-2194. doi:10.21037/tau.2020.03.14
  6. International Germ Cell Cancer Collaborative Group. International germ cell consensus classification: a prognostic factor-based staging system for metastatic germ cell cancers. J Clin Oncol. 1997;15(2):594-603. doi:10.1200/JCO.1997.15.2.594
  7. Beyer J, Collette L, Sauvé N, et al. Survival and new prognosticators in metastatic seminoma: results from the IGCCCG-Update Consortium. J Clin Oncol. 2021;39(14):1553-1562. doi:10.1200/JCO.20.03292
  8. Gillessen S, Sauvé N, Collette L, et al. Predicting outcomes in men with metastatic nonseminomatous germ cell tumors (NSGCT): results from the IGCCCG Update Consortium. J Clin Oncol. 2021;39(14):1563-1574. doi:10.1200/JCO.20.03296
  9. Gilligan T, Lin DW, Aggarwal R, et al. Testicular cancer, version 2.2020, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2019;17(12):1529-1554. doi:10.6004/jnccn.2019.0058
  10. Heinzelbecker J, Schmidt S, Lackner J, et al. Therapy of clinical stage IIa and IIb seminoma: a systematic review. World J Urol. 2022;40(12):2829-2841. doi:10.1007/s00345-021-03873-5
  11. Oldenburg J, Berney DM, Bokemeyer C, et al. Testicular seminoma and nonseminoma: ESMA-EURACAN Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol. 2022;33(4):362-375. doi:10.1016/j.annonc.2022.01.002
  12.  Wymer KM, Pearce SM, Harris KT, Pierorazio PM, Daneshmand S, Eggener SE. Adherence to National Comprehensive Cancer Network® guidelines for testicular cancer. J Urol. 2017;197(3 pt 1):684-689. doi:10.1016/j.juro.2016.09.073
  13. Saoud RM, Andolfi C, Aizen J, et al. Impact of non-guideline-directed care on quality of life in testicular cancer survivors. Eur Urol Focus. 2021;7(5):1137-1142. doi:10.1016/j.euf.2020.10.005
  14. Fung C, Dinh PC, Fossa SD, Travis LB. Testicular cancer survivorship. J Natl Compr Canc Netw. 2019;17(12):1557-1568. doi:10.6004/jnccn.2019.7369
  15. Guo CC, Czerniak B. Somatic-type malignancies in testicular germ cell tumors. Hum Pathol. 2022;127:123-135.
  16. Sprauten M, Darrah TH, Peterson DR, et al. Impact of long-term serum platinum concentrations on neuro- and ototoxicity in cisplatin-treated survivors of testicular cancer. J Clin Oncol. 2012;30(3):300-307. doi:10.1200/JCO.2011.37.4025
  17. Shrem NS, Wood L, Hamilton RJ, et al. Testicular cancer survivorship: long-term toxicity and management. Can Urol Assoc J. 2022;16(8):257-272. doi:10.5489/cuaj.8009
  18. Hellesnes R, Myklebust TA, Fosså SD, et al. Testicular cancer in the cisplatin era: causes of death and mortality rates in a population-based cohort. J Clin Oncol. 2021;39(32):3561-3573. doi:10.1200/JCO.21.00637
  19. Mercieca-Bebber R, Naher SK, Rincones O, Smith AB, Stockler MR. Patient-reported outcomes associated with treatments for testicular cancer: a systematic review. Patient Relat Outcome Meas. 2021;12:129-171. doi:10.2147/PROM.S242754
  20. Sprauten M, Haugnes HS, Brydøy M, et al. Chronic fatigue in 812 testicular cancer survivors during long-term follow-up: increasing prevalence and risk factors. Ann Oncol. 2015;26(10):2133-2140. doi:10.1093/annonc/mdv328
  21. King J, Adra N, Einhorn LH. Testicular cancer: biology to bedside. Cancer Res. 2021;81(21):5369-5376. doi:10.1158/0008-5472.CAN-21-1452
  22. Hiester A, Che Y, Lusch A, et al. Phase 2 single-arm trial of primary retroperitoneal lymph node dissection in patients with seminomatous testicular germ cell tumors with clinical stage IIA/B (PRIMETEST). Eur Urol. 2022;S0302-2838(22)02775-0. doi:10.1016/j.eururo.2022.10.021
  23. Daneshmand S, Cary C, Masterson TA, et al. SEMS trial: result of a prospective, multi-institutional phase II clinical trial of surgery in early metastatic seminoma. J Clin Oncol. 2021;39(6 suppl):Abstract 375. doi:10.1200JCO.2021.39.6_suppl.375
  24. Papachristofilou A, Bedke J, Hayoz S, et al. Single-dose carboplatin followed by involved-node radiotherapy for stage IIA and stage IIB seminoma (SAKK 01/10): a single-arm, multicentre, phase 2 trial. Lancet Oncol. 2022;23(11):1441-1450. doi:10.1016/S1470-2045(22)00564-2
  25. Dieckmann KP, Richter-Simonsen H, Kulejewski M, et al. Testicular germ-cell tumours: a descriptive analysis of clinical characteristics at first presentation. Urol Int. 2018;100(4):409-419. doi:10.1159/000488284
  26. Murray MJ, Huddart RA, Coleman N. The present and future of serum diagnostic tests for testicular germ cell tumours. Nat Rev Urol. 2016;13(12):715-725. doi:10.1038/nrurol.2016.170
Publications
Publications
Topics
Article Type
Display Headline
Progress in Treating Testicular Cancer
Display Headline
Progress in Treating Testicular Cancer
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Mon, 06/19/2023 - 17:45
Un-Gate On Date
Mon, 06/19/2023 - 17:45
Use ProPublica
CFC Schedule Remove Status
Mon, 06/19/2023 - 17:45
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Gastrointestinal Stromal Tumor: Reflecting on 2 Decades of Clinical Advancements

Article Type
Changed
Mon, 06/26/2023 - 12:24
Display Headline
Gastrointestinal Stromal Tumor: Reflecting on 2 Decades of Clinical Advancements

Jason K. Sicklick, MD, FACS
Gastrointestinal stromal tumor (GIST) was recognized as a distinct tumor type in the late 1990s.1 Advances in treatment have expanded since the 2001 US Food and Drug Administration (FDA) approval of imatinib, the first tyrosine kinase inhibitor (TKI).2 In 2023, there are now 5 FDA-approved agents for GIST, and 4 additional agents have been approved (tumor agnostic) for patients whose cancer harbors specific genomic alterations (neurotrophic tyrosine receptor kinase [NTRK] fusions3,4 or BRAF V600E mutations5). According to the National Comprehensive Cancer Network (NCCN) Clinical Practice Guidelines in Oncology, several other drugs (ie, in addition to those specifically approved for GIST) are listed as “useful in certain circumstances.”6

Since the early 2000s, new discoveries about GIST genomics have contributed to better, more targeted treatments. Some genomic mutations have been linked to specific gut regions,which may further help guide therapy as well.

GIST: What Is It, Who Gets It, and How Is It Diagnosed?

What, How Many, Where?

Even though GIST is considered rare—representing less than 1% of gastrointestinal tumors8—it is the most common sarcoma, which is a family of mesenchymal neoplasms. GISTs are thought to arise from the interstitial cells of Cajal, or the pacemaker cells of the gut that control peristalsis. In the United States, the incidence of GIST is roughly 4,000 to 6,000 new cases diagnosed per year, with most cases found in the stomach (60%) or small intestine (35%). Other gut regions in which GISTs may be identified include the rectum and esophagus.8

Although asymptomatic tumors are often discovered incidentally, GISTs that originate in the stomach—the most common primary tumor site—may present with nonspecific subjective symptoms such as pain, nausea, loss of appetite, early satiety, or bloating.9 Symptoms may vary according to tumor location (eg, stomach vs rectum vs esophagus), size, and pattern of growth. More objective signs could include anemia related to gastrointestinal bleeding, weight loss, or a palpable mass.9

Who?

Most cases of GIST occur in patients later in life, with a median age of 64 years at diagnosis. A slight predominance of men has been noted, along with African American and Asian individuals affected somewhat more frequently than White or Hispanic populations.10 GIST is rare in children and adolescents, and the symptoms and pathology differ from those in most adults.Previously age was considered a determining factor in the differences in GIST, with cases in children classified as “pediatric-type” GIST or “wild-type” GIST. These cases generally present in the stomach, are more likely to include lymph node involvement, and can also spread to the liver and abdominal lining. Importantly, they are usually not associated with the tyrosine-protein kinase (KIT) or platelet-derived growth factor receptor alpha (PDGFRA) gene mutations found in most adults.9 About 80% of these cases have hereditary mutations of the succinate dehydrogenase (SDH) enzyme complex. Because some adult cases of GIST share the distinct characteristics found in most pediatric cases, distinguishing them based on age, rather than on the specific genetic characteristics of the tumor, is unwarranted.9

How?

When GIST is suspected or when symptoms mandate further investigation, coordination among colleagues in imaging, gastroenterology, pathology, surgery, and oncology is critical for accurate diagnosis, staging, and treatment. Abdominal imaging may be ordered using modalities such as ultrasound, computed tomography, magnetic resonance imaging, and, occasionally, positron emission tomography.11 Endoscopic ultrasound is useful to identify and biopsy lesions in the stomach or rectum, as these tumors arise below the lining of the stomach or rectum. GIST diagnosis can be confirmed by biopsy during endoscopic ultrasound, which is the preferred approach, or by percutaneous biopsy when endoscopic biopsy is not feasible or safe.11

According to the European Society for Medical Oncology (ESMO) and European Reference Group for Rare Adult Solid Cancers (EURACAN) Clinical Practice Guidelines, the “standard approach to tumors ≥ 2 cm in size is excision, because they are associated with a higher risk of progression if confirmed as GIST. If there is an abdominal nodule not amenable to endoscopic assessment, laparoscopic or laparotomic excision is the standard approach.”11

Genetic Mutations

A diagnosis of GIST is made based on the combination of the clinical scenario, the tumor’s anatomic location, immunohistochemistry patterns, and molecular features.12 Research has shown that genetic mutations in the KIT, PDGFRA, or SDH genes are present in most cases of GIST (70-80%,13 10%,12 and less than 10% of cases12,14 respectively) and their presence can be used for diagnosis. A growing number of rarer mutations have also been discovered,13 meaning that gene-based diagnosis of GIST is becoming increasingly sensitive. In addition, antigens on the surface of cancer cells can help classify them as GIST. For example, researchers have discovered that most GIST cells have the marker CD117, the protein product of the KIT gene that is commonly mutated in GIST, on their surfaces. A different marker, DOG1 (ie, Discovered On GIST 1), is also present on the vast majority of GISTs, but not unanimously overlapping with CD117. A tumor that is positive for both CD117 and DOG1 has a high probability (> 97%) of being GIST.15

Next-generation sequencing (NGS) is considered the best tool for determining both germline and somatic mutations in patients with GIST, and NGS is recommended by both the NCCN6 and the ESMO11 for individualizing systemic therapy. Despite these recommendations, most patients do not undergo genetic testing, both in the United States16 and internationally.17 Several barriers to genetic testing have been cited, predominately inadequate tissue and high cost. However, a study demonstrated that costs of up to $3,730 for genetic testing were ultimately cost-effective for tailoring therapy with first-line imatinib for patients with newly diagnosed metastatic GIST.18 Moreover, genetic testing also should be strongly considered for patients with nonmetastatic disease in whom systemic therapy is being considered.

Increasing evidence has emerged that gastric GIST mutations are related to tumor location within the gastrointestinal tract (Figure).7 The anatomic location of the GIST may provide clues for clinical decision-making and may guide selective confirmatory genomic testing when access to testing is limited.

Proximal gastric GISTs are overwhelmingly KIT exon 11 mutant, whereas distal stomach tumors display non-KIT genomic diversity (PDGFRA, as well as germline [or inherited] SDHx mutant and SDHC epimutations). In contrast, tumors that arise in the small intestine (eg, duodenum, jejunum, and ileum) are associated with gene fusions and germline NF1, as well as mutations in KIT exons 11 and 9. Colon and rectal tumors are likely to be KIT-related, whereas those found in the duodenal-jejunal flexure (also called the ligament of Treitz) correlate with rarer mutations, such as BRAF V600E and somatic (nonhereditary NF1) in addition to germline NF, as well as KIT.

Treatment of GIST

Surgery 

Surgery remains the main treatment for localized GIST, especially if the tumor is discovered at an early stage. Unfortunately, up to a quarter of patients present with metastatic disease at diagnosis. The goal of surgery is to resect the tumor with histologically negative margins. Every effort should be made to avoid rupturing the tumor capsule during resection. Studies have shown that laparoscopic resection is feasible and safe for gastric GISTs and is less invasive than traditional open surgery, with similar oncological outcomes.19

Debulking surgery is sometimes considered for patients with metastatic disease, especially for patients who demonstrate sensitivity to TKI and whose disease has not yet progressed.20,21 Other interventions, such as microwave ablation or transhepatic arterial embolization, are sometimes used to control hepatic metastases.

Systemic Therapies

Whether systemic therapy is being considered in the neoadjuvant (preoperative), adjuvant (postoperative), or advanced disease setting, mutations in GIST determine the likelihood of treatment success. Both NCCN6 and ESMO11 strongly encourage use of mutational analyses and genetic testing for patients with GIST before systemic therapy is initiated.

In some cases of locally advanced GIST, tumors may be situated in particularly challenging anatomic locations(eg, esophagus, duodenum, rectum) or may require a highly morbid, multivisceral resection. In such situations, neoadjuvant treatment with imatinib,22,23 if deemed appropriate per mutational profiling, should be considered.

Patients who are determined to be at high risk for recurrence after surgery, based on tumor size, mitotic index determined by pathologist review of dividing cells, tumor location, and tumor rupture, may be eligible for adjuvant treatment with imatinib.24 Although the ideal duration of adjuvant therapy is not yet known, the current standard is at least 3 years,25 but many practitioners advocate for lifelong therapy.

Imatinib. Because chemotherapy was ineffective against GIST, prognosis was dismal for patients diagnosed with advanced disease before the approval of imatinib in the early 2000s.2 A selective TKI, imatinib targets the KIT and PDGFRA receptor kinases, and most patients experience clinical benefit,26 at least initially. Unfortunately, many tumors eventually develop resistance, and discontinuation of imatinib is associated with a risk for disease progression.27

Sunitinib. The emergence of resistance to imatinib spurred the search for second-line agents that might be useful after disease progression. Another TKI, sunitinib, which has both antitumor and antiangiogenic activity, was approved in 2006 for management of advanced imatinib-resistant GIST.28 Knowledge of a tumor’s driver mutation(s) can help optimize use of sunitinib.29

Regorafenib. In 2013, the FDA approved regorafenib, another TKI, as a third-line agent for patients with advanced GIST that is refractory to imatinib and sunitinib.30 Regorafenib exerts its activity against multiple targets, including VEGFR1-3, TIE2 (ie, antiangiogenic activity), PDGFR-β, FGFR (ie, stromal targets), and KIT, RET, and RAF (ie, oncogenic targets). As with other TKIs, common adverse effects associated with regorafenib treatment include hypertension, hand-foot skin reaction, rash, diarrhea, and fatigue.

Larotrectinib/entrectinib. The FDA approved larotrectinib31 (2018) and entrectinib32 (2019) as the first tumor-agnostic agents, whose use is based on the presence of  specific genomic alteration, in this case NTRK. If a tumor, including a GIST, harbors a specific, albeit rare, gene fusion, it may be considered for treatment with one of these small-molecule TRK family inhibitors. Although these agents are not specifically indicated for GIST, some subjects enrolled in the trials had GIST harboring the target NTRK gene fusion and their tumors responded to treatment.

Ripretinib. FDA-approved in 2020,33 ripretinib is a novel TKI indicated for adult patients with advanced GIST who have received prior treatment with 3 or more kinase inhibitors, including imatinib. A phase 3 trial demonstrated improved progression-free and overall survival when ripretinib was compared with placebo in patients who had disease progression after treatment with imatinib, sunitinib, or regorafenib.34 Ripretinib is now being investigated in the second-line setting in selected patients with KIT mutations.

Avapritinib. Cases of GIST with PDGFRA D842V-mutant tumors often demonstrate primary resistance to imatinib and sunitinib. In 2020, avapritinib, a selective TKI that targets both KIT and PDGFRA, was approved35 for treatment of patients with unresectable or metastatic GIST harboring a PDGFRA exon 18 mutation, including D842V mutations. However, it is noteworthy that many of the non-D842V mutations in PDGFRA respond to imatinib.

Dabrafenib/trametinib. In 2022, the FDA issued an approval for treatment based on a driver mutation rather than tumor type. Acknowledging that a BRAF mutation—specifically a V600E mutation—appears to be a critical target in several cancers, the FDA granted accelerated approval for the use of dabrafenib plus trametinib in adults and children 6 years of age and older with unresectable or metastatic solid tumors with BRAF V600E mutation who have progressed following prior treatment and have no satisfactory alternative treatment options.36

Researchers have studied additional TKIs in the setting of unresectable, metastatic disease due to the varied genomic landscape of GIST. The NCCN Guidelines include evidence of some benefit for agents such as dasatinib, cabozantinib, everolimus (plus a TKI), nilotinib, pazopanib, and sorafenib “in certain circumstances.”6

The Future of GIST

The most critical step toward optimal treatment decision-making when a patient has been diagnosed with GIST is identification of physicians with expertise in the care of patients with GIST. With increasing knowledge of genomic variations in GIST, patient care has become less prescripted and much more personalized. To that end, determination of the tumor’s genetic mutational profile is critical to guiding treatment. Although factors such as cost, availability/accessibility, and insufficient tissue continue to represent substantial obstacles, pursing this information may be the most important way that clinicians can advocate for their patients. Moreover, now that the anatomic location of GIST has been linked to specific driver mutations, the ability to select and refine treatments may improve significantly.

Likewise, in view of the increasing complexity and multidisciplinary management of patients with GIST, efficient coordination is paramount among surgical and medical oncologists, as well as radiologists, gastroenterologists, and pathologists.

Click to read more from 2023 Rare Diseases Report: Cancers

References
  1. Miettinen M, Virolainen M, Maarit-Sarlomo-Rikala. Gastrointestinal stromal tumors—value of CD34 antigen in their identification and separation from true leiomyomas and schwannomas. Am J Surg Pathol. 1995;19(2):207-216. doi:10.1097/00000478-199502000-00009
  2. Dagher R, Cohen M, Williams G, et al. Approval summary: imatinib mesylate in the treatment of metastatic and/or unresectable malignant gastrointestinal stromal tumors. Clin Cancer Res. 2002;8(10):3034-3038. PMID:12374669
  3. US Food and Drug Administration. FDA approves larotrectinib for solid tumors with NTRK gene fusions [press release]. Published November 26, 2018. Accessed March 21, 2023. https://www.fda.gov/drugs/fda-approves-larotrectinib-solid-tumors-ntrk-gene-fusions
  4. US Food and Drug Administration. FDA approves entrectinib for NTRK solid tumors and ROS-1 NSCLC [press release]. Published August 15, 2019. Accessed March 21, 2023. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-entrectinib-ntrk-solid-tumors-and-ros-1-nsclc
  5. Winstead W. Dabrafenib-trametinib combination approved for solid tumors with BRAF mutations. National Institutes of Health, National Cancer Institute. Published July 21, 2022. Accessed March 21, 2023. https://www.cancer.gov/news-events/cancer-currents-blog/2022/fda-dabrafenib-trametinib-braf-solid-tumors
  6. National Comprehensive Cancer Network. NCCN clinic practice guidelines in oncology: gastrointestinal stromal tumors. Version 1.2023. March 13, 2023. Accessed March 21, 2023. https://www.nccn.org/professionals/physician_gls/pdf/gist.pdf
  7. Sharma AK, de la Torre J, IJzerman NS, et al. Location of gastrointestinal stromal tumor (GIST) in the stomach predicts tumor mutation profile and drug sensitivity. Clin Cancer Res. 2021;27(19):5334-5342. doi:10.1158/1078-0432.CCR-21-1221
  8. Gastrointestinal stromal tumor—GIST: statistics. Cancer.net. Published March 2023. Accessed March 21, 2023. https://www.cancer.net/cancer-types/gastrointestinal-stromal-tumor-gist/statistics
  9. Sicklick J. Gastrointestinal stromal tumors - symptoms, causes, treatment: NORD. National Organization for Rare Disorders. Published January 12, 2023. Accessed March 28, 2023. https://rarediseases.org/rare-diseases/gastrointestinal-stromal-tumors/
  10. Ma GL, Murphy JD, Martinez ME, Sicklick JK. Epidemiology of gastrointestinal stromal tumors in the era of histology codes: results of a population-based study. Cancer Epidemiol Biomarkers Prev. 2014;24(1):298-302. doi:10.1158/1055-9965.EPI-14-1002
  11. Casali PG, Abecassis N, Aro HT, et al. Gastrointestinal stromal tumours: ESMOEURACAN Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2018;29(suppl 4):iv68-iv78. doi:10.1093/annonc/mdy095
  12. Kelly CM, Sainz LG, Chi P. The management of metastatic GIST: current standard and investigational therapeutics. J Hematol Oncol. 2021;14(1):2. doi:10.1186/s13045-020-01026-6
  13. Shi E, Chmielecki J, Tang CM, et al. FGR1 and NTRK3 actionable alterations in “wild-type” gastrointestinal stromal tumors. J Translat Med. 2016;14(1):339. doi:10.1186/s12967-016-1075-6
  14. Bannon AE, Klug LR, Corless CL, Heinrich MC. Using molecular diagnostic testing to personalize the treatment of patients with gastrointestinal stromal tumors. Expert Rev Mol Diagn. 2017;17(5):445-457. doi:10.1080/14737159.2017.1308826
  15. Wu CE, Tzen CY, Wang SY, Yeh CN. Clinical diagnosis of gastrointestinal stromal tumor (GIST): from the molecular genetic point of view. Cancers (Basel). 2019;11(5):679. doi:10.3390/cancers11050679
  16. Florindez J, Trent J. Low frequency of mutation testing in the United States: an analysis of 3866 GIST patients. Am J Clin Oncol. 2020;43(4):270-278. doi:10.1097/COC.0000000000000659
  17. Verschoor AJ, Bovée JVMG, Overbeek LIH, PALGA group; Hogendoorn PCW, Gelderblom H. The incidence, mutational status, risk classification and referral pattern of gastrointestinal stromal tumours in the Netherlands: a nationwide pathology registry (PALGA) study. Virchows Arch. 2018;472(2):221-229. doi:10.1007/s00428-017-2285-x
  18. Banerjee S, Kumar A, Lopez N, et al. Cost-effectiveness analysis of genetic testing and tailored first-line therapy for patients with metastatic gastrointestinal stromal tumors. JAMA Netw Open. 2020;3(9):e2013565. doi:10.1001/jamanetworkopen.2020.1356519
  19. Chen K, Zhou YC, Mou YP, Xu XW, Jin WW, Ajoodhea H. Systematic review and meta-analysis of safety and efficacy of laparoscopic resection of gastrointestinal stromal tumors of the stomach. Surg Endosc. 2015;29:355-367. doi:10.1007/s00464-014-3676-6
  20. Fairweather M, Balachandran VP, Li GZ, et al. Cytoreductive surgery for metastatic gastrointestinal stromal tumors treated with tyrosine kinase inhibitors: a 2-institutional analysis. Ann Surg. 2018;268(2):296-302. doi:10.1097/SLA.0000000000002281
  21. Bauer S, Rutkowski P, Hohenberger P, et al. Long-term follow-up of patients with GIST undergoing metastasectomy in the era of imatinib—analysis of prognostic factors (EORTC-STBSG collaborative study). Eur J Surg Oncol. 2014;40(4):412-419. doi:10.1016/j.ejso.2013.12.020
  22. Rutkowski P, Gronchi A, Hohenberger P, et al. Neoadjuvant imatinib in locally advanced gastrointestinal stromal tumors (GIST): the EORTC STBSG experience. Ann Surg Oncol. 2013;20(9):2937-2943. doi:10.1245/s10434-013-3013-7
  23. Cavnar MJ, Seier K, Gönen M, et al. Prognostic factors after neoadjuvant imatinib for newly diagnosed primary gastrointestinal stromal tumor. J Gastrointest Surg. 2021;25(7):1828-1836. doi:10.1007/s11605-020-04843-9
  24. Joensuu H. Risk stratification of patients diagnosed with gastrointestinal stromal tumor. Hum Pathol. 2008;39(10):1411-1419. doi:10.1016/j.humpath.2008.06.025
  25. Joensuu H, Eriksson M, Sundby Hall K, et al. Survival outcomes associated with 3 years vs 1 year of adjuvant imatinib for patients with high-risk gastrointestinal stromal tumors: an analysis of a randomized clinical trial after 10-year follow-up. JAMA Oncol. 2020;6(8):1241-1246. doi:10.1001/jamaoncol.2020.2091
  26. Heinrich MC, Rankin C, Blanke CD, et al. Correlation of long-term results of imatinib in advanced gastrointestinal stromal tumors with next-generation sequencing results: analysis of phase 3 SWOG Intergroup Trial S0033. JAMA Oncol. 2017;3(7):944-952. doi:10.1001/jamaoncol.2016.6728
  27. Le Cesne A, Ray-Coquard I, Bui BN, et al. Discontinuation of imatinib in patients with advanced gastrointestinal stromal tumours after 3 years of treatment: an open-label multicentre randomised phase 3 trial. Lancet Oncol. 2020;11(10):942-949. doi:10.1016/S1470-2045(10)70222-9
  28. Demetri GD, van Oosterom AT, Garrett CR, et al. Efficacy and safety of sunitinib in patients with advanced gastrointestinal stromal tumour after failure of imatinib: a randomised controlled trial. Lancet. 2006;368(9544):1329-1338. doi:10.1016/S0140-6736(06)69446-4
  29. Heinrich MC, Maki RG, Corless CL, et al. Primary and secondary kinase genotypes correlate with the biological and clinical activity of sunitinib in imatinib-resistant gastrointestinal stromal tumor. J Clin Oncol. 2008;26(33):5352-5359. doi:10.1200/JCO.2007.15.7461
  30. Crona DJ, Keisler MD, Walko CM. Regorafenib: a novel multitargeted tyrosine kinase inhibitor for colorectal and gastrointestinal stromal tumors. Ann Pharmacother. 2013;47(12):1685-1696. doi:10.1177/1060028013509792
  31. Drilon A, Laetsch TW, Kummar S, et al. Efficacy of larotrectinib in TRK fusion-positive cancers in adults and children. N Engl J Med. 2018;378(8):731-739. doi:10.1056/NEJMoa1714448
  32. Doebele RC, Drilon A, Paz-Ares L, et al. Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: integrated analysis of three phase 1-2 trials. Lancet Oncol. 2020;21(2):271-282. doi:10.1016/S1470-2045(19)30691-6
  33. US Food and Drug Administration. FDA approves ripretinib for advanced gastrointestinal stromal tumor [press release]. Published May 15, 2020. Accessed March 21, 2023. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-ripretinib-advanced-gastrointestinal-stromal-tumor
  34. Blay JY, Serrano C, Heinrich MC, et al. Ripretinib in patients with advanced gastrointestinal stromal tumours (INVICTUS): A double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol. 2020;21(7):923-934. doi:10.1016/S1470-2045(20)30168-6
  35. US Food and Drug Administration. FDA approves avapritinib for gastrointestinal stromal tumor with a rare mutation [press release]. Published January 9, 2020. Accessed March 21, 2023. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-avapritinib-gastrointestinal-stromal-tumor-rare-mutation
  36. US Food and Drug Administration. FDA grants accelerated approval to dabrafenib in combination with trametinib for unresectable or metastatic solid tumors with BRAF V600E mutation [press release]. Published June 22, 2022. Accessed March 21, 2023. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-dabrafenib-combination-trametinib-unresectable-or-metastatic-solid
     

 

Author and Disclosure Information

Jason K. Sicklick, MD, FACS
Professor of Surgery, Division of Surgical Oncology
Adjunct Professor, Department of Pharmacology, UC San Diego School of Medicine
Executive Vice Chair of Research, Department of Surgery
Associate Program Director, General Surgery Residency Program
Leader, Sarcoma Disease Team, Moores Cancer Center, UC San Diego Health
Co-Leader, Structural and Functional Genomics Program, Moores Cancer Center
La Jolla, CA

Jason K. Sicklick, MD, has disclosed the following relevant financial relationships:

Receives consultant fees from Deciphera; Aadi and Ground Rounds

Serves as a consultant for CureMatch

Received speakers fees from Deciphera; La-Hoffman Roche; Foundation Medicine; Merck; QED; Daiichi Sankyo

Owns stock in Personalis

Publications
Topics
Author and Disclosure Information

Jason K. Sicklick, MD, FACS
Professor of Surgery, Division of Surgical Oncology
Adjunct Professor, Department of Pharmacology, UC San Diego School of Medicine
Executive Vice Chair of Research, Department of Surgery
Associate Program Director, General Surgery Residency Program
Leader, Sarcoma Disease Team, Moores Cancer Center, UC San Diego Health
Co-Leader, Structural and Functional Genomics Program, Moores Cancer Center
La Jolla, CA

Jason K. Sicklick, MD, has disclosed the following relevant financial relationships:

Receives consultant fees from Deciphera; Aadi and Ground Rounds

Serves as a consultant for CureMatch

Received speakers fees from Deciphera; La-Hoffman Roche; Foundation Medicine; Merck; QED; Daiichi Sankyo

Owns stock in Personalis

Author and Disclosure Information

Jason K. Sicklick, MD, FACS
Professor of Surgery, Division of Surgical Oncology
Adjunct Professor, Department of Pharmacology, UC San Diego School of Medicine
Executive Vice Chair of Research, Department of Surgery
Associate Program Director, General Surgery Residency Program
Leader, Sarcoma Disease Team, Moores Cancer Center, UC San Diego Health
Co-Leader, Structural and Functional Genomics Program, Moores Cancer Center
La Jolla, CA

Jason K. Sicklick, MD, has disclosed the following relevant financial relationships:

Receives consultant fees from Deciphera; Aadi and Ground Rounds

Serves as a consultant for CureMatch

Received speakers fees from Deciphera; La-Hoffman Roche; Foundation Medicine; Merck; QED; Daiichi Sankyo

Owns stock in Personalis

Jason K. Sicklick, MD, FACS
Gastrointestinal stromal tumor (GIST) was recognized as a distinct tumor type in the late 1990s.1 Advances in treatment have expanded since the 2001 US Food and Drug Administration (FDA) approval of imatinib, the first tyrosine kinase inhibitor (TKI).2 In 2023, there are now 5 FDA-approved agents for GIST, and 4 additional agents have been approved (tumor agnostic) for patients whose cancer harbors specific genomic alterations (neurotrophic tyrosine receptor kinase [NTRK] fusions3,4 or BRAF V600E mutations5). According to the National Comprehensive Cancer Network (NCCN) Clinical Practice Guidelines in Oncology, several other drugs (ie, in addition to those specifically approved for GIST) are listed as “useful in certain circumstances.”6

Since the early 2000s, new discoveries about GIST genomics have contributed to better, more targeted treatments. Some genomic mutations have been linked to specific gut regions,which may further help guide therapy as well.

GIST: What Is It, Who Gets It, and How Is It Diagnosed?

What, How Many, Where?

Even though GIST is considered rare—representing less than 1% of gastrointestinal tumors8—it is the most common sarcoma, which is a family of mesenchymal neoplasms. GISTs are thought to arise from the interstitial cells of Cajal, or the pacemaker cells of the gut that control peristalsis. In the United States, the incidence of GIST is roughly 4,000 to 6,000 new cases diagnosed per year, with most cases found in the stomach (60%) or small intestine (35%). Other gut regions in which GISTs may be identified include the rectum and esophagus.8

Although asymptomatic tumors are often discovered incidentally, GISTs that originate in the stomach—the most common primary tumor site—may present with nonspecific subjective symptoms such as pain, nausea, loss of appetite, early satiety, or bloating.9 Symptoms may vary according to tumor location (eg, stomach vs rectum vs esophagus), size, and pattern of growth. More objective signs could include anemia related to gastrointestinal bleeding, weight loss, or a palpable mass.9

Who?

Most cases of GIST occur in patients later in life, with a median age of 64 years at diagnosis. A slight predominance of men has been noted, along with African American and Asian individuals affected somewhat more frequently than White or Hispanic populations.10 GIST is rare in children and adolescents, and the symptoms and pathology differ from those in most adults.Previously age was considered a determining factor in the differences in GIST, with cases in children classified as “pediatric-type” GIST or “wild-type” GIST. These cases generally present in the stomach, are more likely to include lymph node involvement, and can also spread to the liver and abdominal lining. Importantly, they are usually not associated with the tyrosine-protein kinase (KIT) or platelet-derived growth factor receptor alpha (PDGFRA) gene mutations found in most adults.9 About 80% of these cases have hereditary mutations of the succinate dehydrogenase (SDH) enzyme complex. Because some adult cases of GIST share the distinct characteristics found in most pediatric cases, distinguishing them based on age, rather than on the specific genetic characteristics of the tumor, is unwarranted.9

How?

When GIST is suspected or when symptoms mandate further investigation, coordination among colleagues in imaging, gastroenterology, pathology, surgery, and oncology is critical for accurate diagnosis, staging, and treatment. Abdominal imaging may be ordered using modalities such as ultrasound, computed tomography, magnetic resonance imaging, and, occasionally, positron emission tomography.11 Endoscopic ultrasound is useful to identify and biopsy lesions in the stomach or rectum, as these tumors arise below the lining of the stomach or rectum. GIST diagnosis can be confirmed by biopsy during endoscopic ultrasound, which is the preferred approach, or by percutaneous biopsy when endoscopic biopsy is not feasible or safe.11

According to the European Society for Medical Oncology (ESMO) and European Reference Group for Rare Adult Solid Cancers (EURACAN) Clinical Practice Guidelines, the “standard approach to tumors ≥ 2 cm in size is excision, because they are associated with a higher risk of progression if confirmed as GIST. If there is an abdominal nodule not amenable to endoscopic assessment, laparoscopic or laparotomic excision is the standard approach.”11

Genetic Mutations

A diagnosis of GIST is made based on the combination of the clinical scenario, the tumor’s anatomic location, immunohistochemistry patterns, and molecular features.12 Research has shown that genetic mutations in the KIT, PDGFRA, or SDH genes are present in most cases of GIST (70-80%,13 10%,12 and less than 10% of cases12,14 respectively) and their presence can be used for diagnosis. A growing number of rarer mutations have also been discovered,13 meaning that gene-based diagnosis of GIST is becoming increasingly sensitive. In addition, antigens on the surface of cancer cells can help classify them as GIST. For example, researchers have discovered that most GIST cells have the marker CD117, the protein product of the KIT gene that is commonly mutated in GIST, on their surfaces. A different marker, DOG1 (ie, Discovered On GIST 1), is also present on the vast majority of GISTs, but not unanimously overlapping with CD117. A tumor that is positive for both CD117 and DOG1 has a high probability (> 97%) of being GIST.15

Next-generation sequencing (NGS) is considered the best tool for determining both germline and somatic mutations in patients with GIST, and NGS is recommended by both the NCCN6 and the ESMO11 for individualizing systemic therapy. Despite these recommendations, most patients do not undergo genetic testing, both in the United States16 and internationally.17 Several barriers to genetic testing have been cited, predominately inadequate tissue and high cost. However, a study demonstrated that costs of up to $3,730 for genetic testing were ultimately cost-effective for tailoring therapy with first-line imatinib for patients with newly diagnosed metastatic GIST.18 Moreover, genetic testing also should be strongly considered for patients with nonmetastatic disease in whom systemic therapy is being considered.

Increasing evidence has emerged that gastric GIST mutations are related to tumor location within the gastrointestinal tract (Figure).7 The anatomic location of the GIST may provide clues for clinical decision-making and may guide selective confirmatory genomic testing when access to testing is limited.

Proximal gastric GISTs are overwhelmingly KIT exon 11 mutant, whereas distal stomach tumors display non-KIT genomic diversity (PDGFRA, as well as germline [or inherited] SDHx mutant and SDHC epimutations). In contrast, tumors that arise in the small intestine (eg, duodenum, jejunum, and ileum) are associated with gene fusions and germline NF1, as well as mutations in KIT exons 11 and 9. Colon and rectal tumors are likely to be KIT-related, whereas those found in the duodenal-jejunal flexure (also called the ligament of Treitz) correlate with rarer mutations, such as BRAF V600E and somatic (nonhereditary NF1) in addition to germline NF, as well as KIT.

Treatment of GIST

Surgery 

Surgery remains the main treatment for localized GIST, especially if the tumor is discovered at an early stage. Unfortunately, up to a quarter of patients present with metastatic disease at diagnosis. The goal of surgery is to resect the tumor with histologically negative margins. Every effort should be made to avoid rupturing the tumor capsule during resection. Studies have shown that laparoscopic resection is feasible and safe for gastric GISTs and is less invasive than traditional open surgery, with similar oncological outcomes.19

Debulking surgery is sometimes considered for patients with metastatic disease, especially for patients who demonstrate sensitivity to TKI and whose disease has not yet progressed.20,21 Other interventions, such as microwave ablation or transhepatic arterial embolization, are sometimes used to control hepatic metastases.

Systemic Therapies

Whether systemic therapy is being considered in the neoadjuvant (preoperative), adjuvant (postoperative), or advanced disease setting, mutations in GIST determine the likelihood of treatment success. Both NCCN6 and ESMO11 strongly encourage use of mutational analyses and genetic testing for patients with GIST before systemic therapy is initiated.

In some cases of locally advanced GIST, tumors may be situated in particularly challenging anatomic locations(eg, esophagus, duodenum, rectum) or may require a highly morbid, multivisceral resection. In such situations, neoadjuvant treatment with imatinib,22,23 if deemed appropriate per mutational profiling, should be considered.

Patients who are determined to be at high risk for recurrence after surgery, based on tumor size, mitotic index determined by pathologist review of dividing cells, tumor location, and tumor rupture, may be eligible for adjuvant treatment with imatinib.24 Although the ideal duration of adjuvant therapy is not yet known, the current standard is at least 3 years,25 but many practitioners advocate for lifelong therapy.

Imatinib. Because chemotherapy was ineffective against GIST, prognosis was dismal for patients diagnosed with advanced disease before the approval of imatinib in the early 2000s.2 A selective TKI, imatinib targets the KIT and PDGFRA receptor kinases, and most patients experience clinical benefit,26 at least initially. Unfortunately, many tumors eventually develop resistance, and discontinuation of imatinib is associated with a risk for disease progression.27

Sunitinib. The emergence of resistance to imatinib spurred the search for second-line agents that might be useful after disease progression. Another TKI, sunitinib, which has both antitumor and antiangiogenic activity, was approved in 2006 for management of advanced imatinib-resistant GIST.28 Knowledge of a tumor’s driver mutation(s) can help optimize use of sunitinib.29

Regorafenib. In 2013, the FDA approved regorafenib, another TKI, as a third-line agent for patients with advanced GIST that is refractory to imatinib and sunitinib.30 Regorafenib exerts its activity against multiple targets, including VEGFR1-3, TIE2 (ie, antiangiogenic activity), PDGFR-β, FGFR (ie, stromal targets), and KIT, RET, and RAF (ie, oncogenic targets). As with other TKIs, common adverse effects associated with regorafenib treatment include hypertension, hand-foot skin reaction, rash, diarrhea, and fatigue.

Larotrectinib/entrectinib. The FDA approved larotrectinib31 (2018) and entrectinib32 (2019) as the first tumor-agnostic agents, whose use is based on the presence of  specific genomic alteration, in this case NTRK. If a tumor, including a GIST, harbors a specific, albeit rare, gene fusion, it may be considered for treatment with one of these small-molecule TRK family inhibitors. Although these agents are not specifically indicated for GIST, some subjects enrolled in the trials had GIST harboring the target NTRK gene fusion and their tumors responded to treatment.

Ripretinib. FDA-approved in 2020,33 ripretinib is a novel TKI indicated for adult patients with advanced GIST who have received prior treatment with 3 or more kinase inhibitors, including imatinib. A phase 3 trial demonstrated improved progression-free and overall survival when ripretinib was compared with placebo in patients who had disease progression after treatment with imatinib, sunitinib, or regorafenib.34 Ripretinib is now being investigated in the second-line setting in selected patients with KIT mutations.

Avapritinib. Cases of GIST with PDGFRA D842V-mutant tumors often demonstrate primary resistance to imatinib and sunitinib. In 2020, avapritinib, a selective TKI that targets both KIT and PDGFRA, was approved35 for treatment of patients with unresectable or metastatic GIST harboring a PDGFRA exon 18 mutation, including D842V mutations. However, it is noteworthy that many of the non-D842V mutations in PDGFRA respond to imatinib.

Dabrafenib/trametinib. In 2022, the FDA issued an approval for treatment based on a driver mutation rather than tumor type. Acknowledging that a BRAF mutation—specifically a V600E mutation—appears to be a critical target in several cancers, the FDA granted accelerated approval for the use of dabrafenib plus trametinib in adults and children 6 years of age and older with unresectable or metastatic solid tumors with BRAF V600E mutation who have progressed following prior treatment and have no satisfactory alternative treatment options.36

Researchers have studied additional TKIs in the setting of unresectable, metastatic disease due to the varied genomic landscape of GIST. The NCCN Guidelines include evidence of some benefit for agents such as dasatinib, cabozantinib, everolimus (plus a TKI), nilotinib, pazopanib, and sorafenib “in certain circumstances.”6

The Future of GIST

The most critical step toward optimal treatment decision-making when a patient has been diagnosed with GIST is identification of physicians with expertise in the care of patients with GIST. With increasing knowledge of genomic variations in GIST, patient care has become less prescripted and much more personalized. To that end, determination of the tumor’s genetic mutational profile is critical to guiding treatment. Although factors such as cost, availability/accessibility, and insufficient tissue continue to represent substantial obstacles, pursing this information may be the most important way that clinicians can advocate for their patients. Moreover, now that the anatomic location of GIST has been linked to specific driver mutations, the ability to select and refine treatments may improve significantly.

Likewise, in view of the increasing complexity and multidisciplinary management of patients with GIST, efficient coordination is paramount among surgical and medical oncologists, as well as radiologists, gastroenterologists, and pathologists.

Click to read more from 2023 Rare Diseases Report: Cancers

Jason K. Sicklick, MD, FACS
Gastrointestinal stromal tumor (GIST) was recognized as a distinct tumor type in the late 1990s.1 Advances in treatment have expanded since the 2001 US Food and Drug Administration (FDA) approval of imatinib, the first tyrosine kinase inhibitor (TKI).2 In 2023, there are now 5 FDA-approved agents for GIST, and 4 additional agents have been approved (tumor agnostic) for patients whose cancer harbors specific genomic alterations (neurotrophic tyrosine receptor kinase [NTRK] fusions3,4 or BRAF V600E mutations5). According to the National Comprehensive Cancer Network (NCCN) Clinical Practice Guidelines in Oncology, several other drugs (ie, in addition to those specifically approved for GIST) are listed as “useful in certain circumstances.”6

Since the early 2000s, new discoveries about GIST genomics have contributed to better, more targeted treatments. Some genomic mutations have been linked to specific gut regions,which may further help guide therapy as well.

GIST: What Is It, Who Gets It, and How Is It Diagnosed?

What, How Many, Where?

Even though GIST is considered rare—representing less than 1% of gastrointestinal tumors8—it is the most common sarcoma, which is a family of mesenchymal neoplasms. GISTs are thought to arise from the interstitial cells of Cajal, or the pacemaker cells of the gut that control peristalsis. In the United States, the incidence of GIST is roughly 4,000 to 6,000 new cases diagnosed per year, with most cases found in the stomach (60%) or small intestine (35%). Other gut regions in which GISTs may be identified include the rectum and esophagus.8

Although asymptomatic tumors are often discovered incidentally, GISTs that originate in the stomach—the most common primary tumor site—may present with nonspecific subjective symptoms such as pain, nausea, loss of appetite, early satiety, or bloating.9 Symptoms may vary according to tumor location (eg, stomach vs rectum vs esophagus), size, and pattern of growth. More objective signs could include anemia related to gastrointestinal bleeding, weight loss, or a palpable mass.9

Who?

Most cases of GIST occur in patients later in life, with a median age of 64 years at diagnosis. A slight predominance of men has been noted, along with African American and Asian individuals affected somewhat more frequently than White or Hispanic populations.10 GIST is rare in children and adolescents, and the symptoms and pathology differ from those in most adults.Previously age was considered a determining factor in the differences in GIST, with cases in children classified as “pediatric-type” GIST or “wild-type” GIST. These cases generally present in the stomach, are more likely to include lymph node involvement, and can also spread to the liver and abdominal lining. Importantly, they are usually not associated with the tyrosine-protein kinase (KIT) or platelet-derived growth factor receptor alpha (PDGFRA) gene mutations found in most adults.9 About 80% of these cases have hereditary mutations of the succinate dehydrogenase (SDH) enzyme complex. Because some adult cases of GIST share the distinct characteristics found in most pediatric cases, distinguishing them based on age, rather than on the specific genetic characteristics of the tumor, is unwarranted.9

How?

When GIST is suspected or when symptoms mandate further investigation, coordination among colleagues in imaging, gastroenterology, pathology, surgery, and oncology is critical for accurate diagnosis, staging, and treatment. Abdominal imaging may be ordered using modalities such as ultrasound, computed tomography, magnetic resonance imaging, and, occasionally, positron emission tomography.11 Endoscopic ultrasound is useful to identify and biopsy lesions in the stomach or rectum, as these tumors arise below the lining of the stomach or rectum. GIST diagnosis can be confirmed by biopsy during endoscopic ultrasound, which is the preferred approach, or by percutaneous biopsy when endoscopic biopsy is not feasible or safe.11

According to the European Society for Medical Oncology (ESMO) and European Reference Group for Rare Adult Solid Cancers (EURACAN) Clinical Practice Guidelines, the “standard approach to tumors ≥ 2 cm in size is excision, because they are associated with a higher risk of progression if confirmed as GIST. If there is an abdominal nodule not amenable to endoscopic assessment, laparoscopic or laparotomic excision is the standard approach.”11

Genetic Mutations

A diagnosis of GIST is made based on the combination of the clinical scenario, the tumor’s anatomic location, immunohistochemistry patterns, and molecular features.12 Research has shown that genetic mutations in the KIT, PDGFRA, or SDH genes are present in most cases of GIST (70-80%,13 10%,12 and less than 10% of cases12,14 respectively) and their presence can be used for diagnosis. A growing number of rarer mutations have also been discovered,13 meaning that gene-based diagnosis of GIST is becoming increasingly sensitive. In addition, antigens on the surface of cancer cells can help classify them as GIST. For example, researchers have discovered that most GIST cells have the marker CD117, the protein product of the KIT gene that is commonly mutated in GIST, on their surfaces. A different marker, DOG1 (ie, Discovered On GIST 1), is also present on the vast majority of GISTs, but not unanimously overlapping with CD117. A tumor that is positive for both CD117 and DOG1 has a high probability (> 97%) of being GIST.15

Next-generation sequencing (NGS) is considered the best tool for determining both germline and somatic mutations in patients with GIST, and NGS is recommended by both the NCCN6 and the ESMO11 for individualizing systemic therapy. Despite these recommendations, most patients do not undergo genetic testing, both in the United States16 and internationally.17 Several barriers to genetic testing have been cited, predominately inadequate tissue and high cost. However, a study demonstrated that costs of up to $3,730 for genetic testing were ultimately cost-effective for tailoring therapy with first-line imatinib for patients with newly diagnosed metastatic GIST.18 Moreover, genetic testing also should be strongly considered for patients with nonmetastatic disease in whom systemic therapy is being considered.

Increasing evidence has emerged that gastric GIST mutations are related to tumor location within the gastrointestinal tract (Figure).7 The anatomic location of the GIST may provide clues for clinical decision-making and may guide selective confirmatory genomic testing when access to testing is limited.

Proximal gastric GISTs are overwhelmingly KIT exon 11 mutant, whereas distal stomach tumors display non-KIT genomic diversity (PDGFRA, as well as germline [or inherited] SDHx mutant and SDHC epimutations). In contrast, tumors that arise in the small intestine (eg, duodenum, jejunum, and ileum) are associated with gene fusions and germline NF1, as well as mutations in KIT exons 11 and 9. Colon and rectal tumors are likely to be KIT-related, whereas those found in the duodenal-jejunal flexure (also called the ligament of Treitz) correlate with rarer mutations, such as BRAF V600E and somatic (nonhereditary NF1) in addition to germline NF, as well as KIT.

Treatment of GIST

Surgery 

Surgery remains the main treatment for localized GIST, especially if the tumor is discovered at an early stage. Unfortunately, up to a quarter of patients present with metastatic disease at diagnosis. The goal of surgery is to resect the tumor with histologically negative margins. Every effort should be made to avoid rupturing the tumor capsule during resection. Studies have shown that laparoscopic resection is feasible and safe for gastric GISTs and is less invasive than traditional open surgery, with similar oncological outcomes.19

Debulking surgery is sometimes considered for patients with metastatic disease, especially for patients who demonstrate sensitivity to TKI and whose disease has not yet progressed.20,21 Other interventions, such as microwave ablation or transhepatic arterial embolization, are sometimes used to control hepatic metastases.

Systemic Therapies

Whether systemic therapy is being considered in the neoadjuvant (preoperative), adjuvant (postoperative), or advanced disease setting, mutations in GIST determine the likelihood of treatment success. Both NCCN6 and ESMO11 strongly encourage use of mutational analyses and genetic testing for patients with GIST before systemic therapy is initiated.

In some cases of locally advanced GIST, tumors may be situated in particularly challenging anatomic locations(eg, esophagus, duodenum, rectum) or may require a highly morbid, multivisceral resection. In such situations, neoadjuvant treatment with imatinib,22,23 if deemed appropriate per mutational profiling, should be considered.

Patients who are determined to be at high risk for recurrence after surgery, based on tumor size, mitotic index determined by pathologist review of dividing cells, tumor location, and tumor rupture, may be eligible for adjuvant treatment with imatinib.24 Although the ideal duration of adjuvant therapy is not yet known, the current standard is at least 3 years,25 but many practitioners advocate for lifelong therapy.

Imatinib. Because chemotherapy was ineffective against GIST, prognosis was dismal for patients diagnosed with advanced disease before the approval of imatinib in the early 2000s.2 A selective TKI, imatinib targets the KIT and PDGFRA receptor kinases, and most patients experience clinical benefit,26 at least initially. Unfortunately, many tumors eventually develop resistance, and discontinuation of imatinib is associated with a risk for disease progression.27

Sunitinib. The emergence of resistance to imatinib spurred the search for second-line agents that might be useful after disease progression. Another TKI, sunitinib, which has both antitumor and antiangiogenic activity, was approved in 2006 for management of advanced imatinib-resistant GIST.28 Knowledge of a tumor’s driver mutation(s) can help optimize use of sunitinib.29

Regorafenib. In 2013, the FDA approved regorafenib, another TKI, as a third-line agent for patients with advanced GIST that is refractory to imatinib and sunitinib.30 Regorafenib exerts its activity against multiple targets, including VEGFR1-3, TIE2 (ie, antiangiogenic activity), PDGFR-β, FGFR (ie, stromal targets), and KIT, RET, and RAF (ie, oncogenic targets). As with other TKIs, common adverse effects associated with regorafenib treatment include hypertension, hand-foot skin reaction, rash, diarrhea, and fatigue.

Larotrectinib/entrectinib. The FDA approved larotrectinib31 (2018) and entrectinib32 (2019) as the first tumor-agnostic agents, whose use is based on the presence of  specific genomic alteration, in this case NTRK. If a tumor, including a GIST, harbors a specific, albeit rare, gene fusion, it may be considered for treatment with one of these small-molecule TRK family inhibitors. Although these agents are not specifically indicated for GIST, some subjects enrolled in the trials had GIST harboring the target NTRK gene fusion and their tumors responded to treatment.

Ripretinib. FDA-approved in 2020,33 ripretinib is a novel TKI indicated for adult patients with advanced GIST who have received prior treatment with 3 or more kinase inhibitors, including imatinib. A phase 3 trial demonstrated improved progression-free and overall survival when ripretinib was compared with placebo in patients who had disease progression after treatment with imatinib, sunitinib, or regorafenib.34 Ripretinib is now being investigated in the second-line setting in selected patients with KIT mutations.

Avapritinib. Cases of GIST with PDGFRA D842V-mutant tumors often demonstrate primary resistance to imatinib and sunitinib. In 2020, avapritinib, a selective TKI that targets both KIT and PDGFRA, was approved35 for treatment of patients with unresectable or metastatic GIST harboring a PDGFRA exon 18 mutation, including D842V mutations. However, it is noteworthy that many of the non-D842V mutations in PDGFRA respond to imatinib.

Dabrafenib/trametinib. In 2022, the FDA issued an approval for treatment based on a driver mutation rather than tumor type. Acknowledging that a BRAF mutation—specifically a V600E mutation—appears to be a critical target in several cancers, the FDA granted accelerated approval for the use of dabrafenib plus trametinib in adults and children 6 years of age and older with unresectable or metastatic solid tumors with BRAF V600E mutation who have progressed following prior treatment and have no satisfactory alternative treatment options.36

Researchers have studied additional TKIs in the setting of unresectable, metastatic disease due to the varied genomic landscape of GIST. The NCCN Guidelines include evidence of some benefit for agents such as dasatinib, cabozantinib, everolimus (plus a TKI), nilotinib, pazopanib, and sorafenib “in certain circumstances.”6

The Future of GIST

The most critical step toward optimal treatment decision-making when a patient has been diagnosed with GIST is identification of physicians with expertise in the care of patients with GIST. With increasing knowledge of genomic variations in GIST, patient care has become less prescripted and much more personalized. To that end, determination of the tumor’s genetic mutational profile is critical to guiding treatment. Although factors such as cost, availability/accessibility, and insufficient tissue continue to represent substantial obstacles, pursing this information may be the most important way that clinicians can advocate for their patients. Moreover, now that the anatomic location of GIST has been linked to specific driver mutations, the ability to select and refine treatments may improve significantly.

Likewise, in view of the increasing complexity and multidisciplinary management of patients with GIST, efficient coordination is paramount among surgical and medical oncologists, as well as radiologists, gastroenterologists, and pathologists.

Click to read more from 2023 Rare Diseases Report: Cancers

References
  1. Miettinen M, Virolainen M, Maarit-Sarlomo-Rikala. Gastrointestinal stromal tumors—value of CD34 antigen in their identification and separation from true leiomyomas and schwannomas. Am J Surg Pathol. 1995;19(2):207-216. doi:10.1097/00000478-199502000-00009
  2. Dagher R, Cohen M, Williams G, et al. Approval summary: imatinib mesylate in the treatment of metastatic and/or unresectable malignant gastrointestinal stromal tumors. Clin Cancer Res. 2002;8(10):3034-3038. PMID:12374669
  3. US Food and Drug Administration. FDA approves larotrectinib for solid tumors with NTRK gene fusions [press release]. Published November 26, 2018. Accessed March 21, 2023. https://www.fda.gov/drugs/fda-approves-larotrectinib-solid-tumors-ntrk-gene-fusions
  4. US Food and Drug Administration. FDA approves entrectinib for NTRK solid tumors and ROS-1 NSCLC [press release]. Published August 15, 2019. Accessed March 21, 2023. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-entrectinib-ntrk-solid-tumors-and-ros-1-nsclc
  5. Winstead W. Dabrafenib-trametinib combination approved for solid tumors with BRAF mutations. National Institutes of Health, National Cancer Institute. Published July 21, 2022. Accessed March 21, 2023. https://www.cancer.gov/news-events/cancer-currents-blog/2022/fda-dabrafenib-trametinib-braf-solid-tumors
  6. National Comprehensive Cancer Network. NCCN clinic practice guidelines in oncology: gastrointestinal stromal tumors. Version 1.2023. March 13, 2023. Accessed March 21, 2023. https://www.nccn.org/professionals/physician_gls/pdf/gist.pdf
  7. Sharma AK, de la Torre J, IJzerman NS, et al. Location of gastrointestinal stromal tumor (GIST) in the stomach predicts tumor mutation profile and drug sensitivity. Clin Cancer Res. 2021;27(19):5334-5342. doi:10.1158/1078-0432.CCR-21-1221
  8. Gastrointestinal stromal tumor—GIST: statistics. Cancer.net. Published March 2023. Accessed March 21, 2023. https://www.cancer.net/cancer-types/gastrointestinal-stromal-tumor-gist/statistics
  9. Sicklick J. Gastrointestinal stromal tumors - symptoms, causes, treatment: NORD. National Organization for Rare Disorders. Published January 12, 2023. Accessed March 28, 2023. https://rarediseases.org/rare-diseases/gastrointestinal-stromal-tumors/
  10. Ma GL, Murphy JD, Martinez ME, Sicklick JK. Epidemiology of gastrointestinal stromal tumors in the era of histology codes: results of a population-based study. Cancer Epidemiol Biomarkers Prev. 2014;24(1):298-302. doi:10.1158/1055-9965.EPI-14-1002
  11. Casali PG, Abecassis N, Aro HT, et al. Gastrointestinal stromal tumours: ESMOEURACAN Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2018;29(suppl 4):iv68-iv78. doi:10.1093/annonc/mdy095
  12. Kelly CM, Sainz LG, Chi P. The management of metastatic GIST: current standard and investigational therapeutics. J Hematol Oncol. 2021;14(1):2. doi:10.1186/s13045-020-01026-6
  13. Shi E, Chmielecki J, Tang CM, et al. FGR1 and NTRK3 actionable alterations in “wild-type” gastrointestinal stromal tumors. J Translat Med. 2016;14(1):339. doi:10.1186/s12967-016-1075-6
  14. Bannon AE, Klug LR, Corless CL, Heinrich MC. Using molecular diagnostic testing to personalize the treatment of patients with gastrointestinal stromal tumors. Expert Rev Mol Diagn. 2017;17(5):445-457. doi:10.1080/14737159.2017.1308826
  15. Wu CE, Tzen CY, Wang SY, Yeh CN. Clinical diagnosis of gastrointestinal stromal tumor (GIST): from the molecular genetic point of view. Cancers (Basel). 2019;11(5):679. doi:10.3390/cancers11050679
  16. Florindez J, Trent J. Low frequency of mutation testing in the United States: an analysis of 3866 GIST patients. Am J Clin Oncol. 2020;43(4):270-278. doi:10.1097/COC.0000000000000659
  17. Verschoor AJ, Bovée JVMG, Overbeek LIH, PALGA group; Hogendoorn PCW, Gelderblom H. The incidence, mutational status, risk classification and referral pattern of gastrointestinal stromal tumours in the Netherlands: a nationwide pathology registry (PALGA) study. Virchows Arch. 2018;472(2):221-229. doi:10.1007/s00428-017-2285-x
  18. Banerjee S, Kumar A, Lopez N, et al. Cost-effectiveness analysis of genetic testing and tailored first-line therapy for patients with metastatic gastrointestinal stromal tumors. JAMA Netw Open. 2020;3(9):e2013565. doi:10.1001/jamanetworkopen.2020.1356519
  19. Chen K, Zhou YC, Mou YP, Xu XW, Jin WW, Ajoodhea H. Systematic review and meta-analysis of safety and efficacy of laparoscopic resection of gastrointestinal stromal tumors of the stomach. Surg Endosc. 2015;29:355-367. doi:10.1007/s00464-014-3676-6
  20. Fairweather M, Balachandran VP, Li GZ, et al. Cytoreductive surgery for metastatic gastrointestinal stromal tumors treated with tyrosine kinase inhibitors: a 2-institutional analysis. Ann Surg. 2018;268(2):296-302. doi:10.1097/SLA.0000000000002281
  21. Bauer S, Rutkowski P, Hohenberger P, et al. Long-term follow-up of patients with GIST undergoing metastasectomy in the era of imatinib—analysis of prognostic factors (EORTC-STBSG collaborative study). Eur J Surg Oncol. 2014;40(4):412-419. doi:10.1016/j.ejso.2013.12.020
  22. Rutkowski P, Gronchi A, Hohenberger P, et al. Neoadjuvant imatinib in locally advanced gastrointestinal stromal tumors (GIST): the EORTC STBSG experience. Ann Surg Oncol. 2013;20(9):2937-2943. doi:10.1245/s10434-013-3013-7
  23. Cavnar MJ, Seier K, Gönen M, et al. Prognostic factors after neoadjuvant imatinib for newly diagnosed primary gastrointestinal stromal tumor. J Gastrointest Surg. 2021;25(7):1828-1836. doi:10.1007/s11605-020-04843-9
  24. Joensuu H. Risk stratification of patients diagnosed with gastrointestinal stromal tumor. Hum Pathol. 2008;39(10):1411-1419. doi:10.1016/j.humpath.2008.06.025
  25. Joensuu H, Eriksson M, Sundby Hall K, et al. Survival outcomes associated with 3 years vs 1 year of adjuvant imatinib for patients with high-risk gastrointestinal stromal tumors: an analysis of a randomized clinical trial after 10-year follow-up. JAMA Oncol. 2020;6(8):1241-1246. doi:10.1001/jamaoncol.2020.2091
  26. Heinrich MC, Rankin C, Blanke CD, et al. Correlation of long-term results of imatinib in advanced gastrointestinal stromal tumors with next-generation sequencing results: analysis of phase 3 SWOG Intergroup Trial S0033. JAMA Oncol. 2017;3(7):944-952. doi:10.1001/jamaoncol.2016.6728
  27. Le Cesne A, Ray-Coquard I, Bui BN, et al. Discontinuation of imatinib in patients with advanced gastrointestinal stromal tumours after 3 years of treatment: an open-label multicentre randomised phase 3 trial. Lancet Oncol. 2020;11(10):942-949. doi:10.1016/S1470-2045(10)70222-9
  28. Demetri GD, van Oosterom AT, Garrett CR, et al. Efficacy and safety of sunitinib in patients with advanced gastrointestinal stromal tumour after failure of imatinib: a randomised controlled trial. Lancet. 2006;368(9544):1329-1338. doi:10.1016/S0140-6736(06)69446-4
  29. Heinrich MC, Maki RG, Corless CL, et al. Primary and secondary kinase genotypes correlate with the biological and clinical activity of sunitinib in imatinib-resistant gastrointestinal stromal tumor. J Clin Oncol. 2008;26(33):5352-5359. doi:10.1200/JCO.2007.15.7461
  30. Crona DJ, Keisler MD, Walko CM. Regorafenib: a novel multitargeted tyrosine kinase inhibitor for colorectal and gastrointestinal stromal tumors. Ann Pharmacother. 2013;47(12):1685-1696. doi:10.1177/1060028013509792
  31. Drilon A, Laetsch TW, Kummar S, et al. Efficacy of larotrectinib in TRK fusion-positive cancers in adults and children. N Engl J Med. 2018;378(8):731-739. doi:10.1056/NEJMoa1714448
  32. Doebele RC, Drilon A, Paz-Ares L, et al. Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: integrated analysis of three phase 1-2 trials. Lancet Oncol. 2020;21(2):271-282. doi:10.1016/S1470-2045(19)30691-6
  33. US Food and Drug Administration. FDA approves ripretinib for advanced gastrointestinal stromal tumor [press release]. Published May 15, 2020. Accessed March 21, 2023. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-ripretinib-advanced-gastrointestinal-stromal-tumor
  34. Blay JY, Serrano C, Heinrich MC, et al. Ripretinib in patients with advanced gastrointestinal stromal tumours (INVICTUS): A double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol. 2020;21(7):923-934. doi:10.1016/S1470-2045(20)30168-6
  35. US Food and Drug Administration. FDA approves avapritinib for gastrointestinal stromal tumor with a rare mutation [press release]. Published January 9, 2020. Accessed March 21, 2023. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-avapritinib-gastrointestinal-stromal-tumor-rare-mutation
  36. US Food and Drug Administration. FDA grants accelerated approval to dabrafenib in combination with trametinib for unresectable or metastatic solid tumors with BRAF V600E mutation [press release]. Published June 22, 2022. Accessed March 21, 2023. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-dabrafenib-combination-trametinib-unresectable-or-metastatic-solid
     

 

References
  1. Miettinen M, Virolainen M, Maarit-Sarlomo-Rikala. Gastrointestinal stromal tumors—value of CD34 antigen in their identification and separation from true leiomyomas and schwannomas. Am J Surg Pathol. 1995;19(2):207-216. doi:10.1097/00000478-199502000-00009
  2. Dagher R, Cohen M, Williams G, et al. Approval summary: imatinib mesylate in the treatment of metastatic and/or unresectable malignant gastrointestinal stromal tumors. Clin Cancer Res. 2002;8(10):3034-3038. PMID:12374669
  3. US Food and Drug Administration. FDA approves larotrectinib for solid tumors with NTRK gene fusions [press release]. Published November 26, 2018. Accessed March 21, 2023. https://www.fda.gov/drugs/fda-approves-larotrectinib-solid-tumors-ntrk-gene-fusions
  4. US Food and Drug Administration. FDA approves entrectinib for NTRK solid tumors and ROS-1 NSCLC [press release]. Published August 15, 2019. Accessed March 21, 2023. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-entrectinib-ntrk-solid-tumors-and-ros-1-nsclc
  5. Winstead W. Dabrafenib-trametinib combination approved for solid tumors with BRAF mutations. National Institutes of Health, National Cancer Institute. Published July 21, 2022. Accessed March 21, 2023. https://www.cancer.gov/news-events/cancer-currents-blog/2022/fda-dabrafenib-trametinib-braf-solid-tumors
  6. National Comprehensive Cancer Network. NCCN clinic practice guidelines in oncology: gastrointestinal stromal tumors. Version 1.2023. March 13, 2023. Accessed March 21, 2023. https://www.nccn.org/professionals/physician_gls/pdf/gist.pdf
  7. Sharma AK, de la Torre J, IJzerman NS, et al. Location of gastrointestinal stromal tumor (GIST) in the stomach predicts tumor mutation profile and drug sensitivity. Clin Cancer Res. 2021;27(19):5334-5342. doi:10.1158/1078-0432.CCR-21-1221
  8. Gastrointestinal stromal tumor—GIST: statistics. Cancer.net. Published March 2023. Accessed March 21, 2023. https://www.cancer.net/cancer-types/gastrointestinal-stromal-tumor-gist/statistics
  9. Sicklick J. Gastrointestinal stromal tumors - symptoms, causes, treatment: NORD. National Organization for Rare Disorders. Published January 12, 2023. Accessed March 28, 2023. https://rarediseases.org/rare-diseases/gastrointestinal-stromal-tumors/
  10. Ma GL, Murphy JD, Martinez ME, Sicklick JK. Epidemiology of gastrointestinal stromal tumors in the era of histology codes: results of a population-based study. Cancer Epidemiol Biomarkers Prev. 2014;24(1):298-302. doi:10.1158/1055-9965.EPI-14-1002
  11. Casali PG, Abecassis N, Aro HT, et al. Gastrointestinal stromal tumours: ESMOEURACAN Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2018;29(suppl 4):iv68-iv78. doi:10.1093/annonc/mdy095
  12. Kelly CM, Sainz LG, Chi P. The management of metastatic GIST: current standard and investigational therapeutics. J Hematol Oncol. 2021;14(1):2. doi:10.1186/s13045-020-01026-6
  13. Shi E, Chmielecki J, Tang CM, et al. FGR1 and NTRK3 actionable alterations in “wild-type” gastrointestinal stromal tumors. J Translat Med. 2016;14(1):339. doi:10.1186/s12967-016-1075-6
  14. Bannon AE, Klug LR, Corless CL, Heinrich MC. Using molecular diagnostic testing to personalize the treatment of patients with gastrointestinal stromal tumors. Expert Rev Mol Diagn. 2017;17(5):445-457. doi:10.1080/14737159.2017.1308826
  15. Wu CE, Tzen CY, Wang SY, Yeh CN. Clinical diagnosis of gastrointestinal stromal tumor (GIST): from the molecular genetic point of view. Cancers (Basel). 2019;11(5):679. doi:10.3390/cancers11050679
  16. Florindez J, Trent J. Low frequency of mutation testing in the United States: an analysis of 3866 GIST patients. Am J Clin Oncol. 2020;43(4):270-278. doi:10.1097/COC.0000000000000659
  17. Verschoor AJ, Bovée JVMG, Overbeek LIH, PALGA group; Hogendoorn PCW, Gelderblom H. The incidence, mutational status, risk classification and referral pattern of gastrointestinal stromal tumours in the Netherlands: a nationwide pathology registry (PALGA) study. Virchows Arch. 2018;472(2):221-229. doi:10.1007/s00428-017-2285-x
  18. Banerjee S, Kumar A, Lopez N, et al. Cost-effectiveness analysis of genetic testing and tailored first-line therapy for patients with metastatic gastrointestinal stromal tumors. JAMA Netw Open. 2020;3(9):e2013565. doi:10.1001/jamanetworkopen.2020.1356519
  19. Chen K, Zhou YC, Mou YP, Xu XW, Jin WW, Ajoodhea H. Systematic review and meta-analysis of safety and efficacy of laparoscopic resection of gastrointestinal stromal tumors of the stomach. Surg Endosc. 2015;29:355-367. doi:10.1007/s00464-014-3676-6
  20. Fairweather M, Balachandran VP, Li GZ, et al. Cytoreductive surgery for metastatic gastrointestinal stromal tumors treated with tyrosine kinase inhibitors: a 2-institutional analysis. Ann Surg. 2018;268(2):296-302. doi:10.1097/SLA.0000000000002281
  21. Bauer S, Rutkowski P, Hohenberger P, et al. Long-term follow-up of patients with GIST undergoing metastasectomy in the era of imatinib—analysis of prognostic factors (EORTC-STBSG collaborative study). Eur J Surg Oncol. 2014;40(4):412-419. doi:10.1016/j.ejso.2013.12.020
  22. Rutkowski P, Gronchi A, Hohenberger P, et al. Neoadjuvant imatinib in locally advanced gastrointestinal stromal tumors (GIST): the EORTC STBSG experience. Ann Surg Oncol. 2013;20(9):2937-2943. doi:10.1245/s10434-013-3013-7
  23. Cavnar MJ, Seier K, Gönen M, et al. Prognostic factors after neoadjuvant imatinib for newly diagnosed primary gastrointestinal stromal tumor. J Gastrointest Surg. 2021;25(7):1828-1836. doi:10.1007/s11605-020-04843-9
  24. Joensuu H. Risk stratification of patients diagnosed with gastrointestinal stromal tumor. Hum Pathol. 2008;39(10):1411-1419. doi:10.1016/j.humpath.2008.06.025
  25. Joensuu H, Eriksson M, Sundby Hall K, et al. Survival outcomes associated with 3 years vs 1 year of adjuvant imatinib for patients with high-risk gastrointestinal stromal tumors: an analysis of a randomized clinical trial after 10-year follow-up. JAMA Oncol. 2020;6(8):1241-1246. doi:10.1001/jamaoncol.2020.2091
  26. Heinrich MC, Rankin C, Blanke CD, et al. Correlation of long-term results of imatinib in advanced gastrointestinal stromal tumors with next-generation sequencing results: analysis of phase 3 SWOG Intergroup Trial S0033. JAMA Oncol. 2017;3(7):944-952. doi:10.1001/jamaoncol.2016.6728
  27. Le Cesne A, Ray-Coquard I, Bui BN, et al. Discontinuation of imatinib in patients with advanced gastrointestinal stromal tumours after 3 years of treatment: an open-label multicentre randomised phase 3 trial. Lancet Oncol. 2020;11(10):942-949. doi:10.1016/S1470-2045(10)70222-9
  28. Demetri GD, van Oosterom AT, Garrett CR, et al. Efficacy and safety of sunitinib in patients with advanced gastrointestinal stromal tumour after failure of imatinib: a randomised controlled trial. Lancet. 2006;368(9544):1329-1338. doi:10.1016/S0140-6736(06)69446-4
  29. Heinrich MC, Maki RG, Corless CL, et al. Primary and secondary kinase genotypes correlate with the biological and clinical activity of sunitinib in imatinib-resistant gastrointestinal stromal tumor. J Clin Oncol. 2008;26(33):5352-5359. doi:10.1200/JCO.2007.15.7461
  30. Crona DJ, Keisler MD, Walko CM. Regorafenib: a novel multitargeted tyrosine kinase inhibitor for colorectal and gastrointestinal stromal tumors. Ann Pharmacother. 2013;47(12):1685-1696. doi:10.1177/1060028013509792
  31. Drilon A, Laetsch TW, Kummar S, et al. Efficacy of larotrectinib in TRK fusion-positive cancers in adults and children. N Engl J Med. 2018;378(8):731-739. doi:10.1056/NEJMoa1714448
  32. Doebele RC, Drilon A, Paz-Ares L, et al. Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: integrated analysis of three phase 1-2 trials. Lancet Oncol. 2020;21(2):271-282. doi:10.1016/S1470-2045(19)30691-6
  33. US Food and Drug Administration. FDA approves ripretinib for advanced gastrointestinal stromal tumor [press release]. Published May 15, 2020. Accessed March 21, 2023. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-ripretinib-advanced-gastrointestinal-stromal-tumor
  34. Blay JY, Serrano C, Heinrich MC, et al. Ripretinib in patients with advanced gastrointestinal stromal tumours (INVICTUS): A double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol. 2020;21(7):923-934. doi:10.1016/S1470-2045(20)30168-6
  35. US Food and Drug Administration. FDA approves avapritinib for gastrointestinal stromal tumor with a rare mutation [press release]. Published January 9, 2020. Accessed March 21, 2023. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-avapritinib-gastrointestinal-stromal-tumor-rare-mutation
  36. US Food and Drug Administration. FDA grants accelerated approval to dabrafenib in combination with trametinib for unresectable or metastatic solid tumors with BRAF V600E mutation [press release]. Published June 22, 2022. Accessed March 21, 2023. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-dabrafenib-combination-trametinib-unresectable-or-metastatic-solid
     

 

Publications
Publications
Topics
Article Type
Display Headline
Gastrointestinal Stromal Tumor: Reflecting on 2 Decades of Clinical Advancements
Display Headline
Gastrointestinal Stromal Tumor: Reflecting on 2 Decades of Clinical Advancements
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Mon, 06/19/2023 - 14:45
Un-Gate On Date
Mon, 06/19/2023 - 14:45
Use ProPublica
CFC Schedule Remove Status
Mon, 06/19/2023 - 14:45
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

An Evolving Understanding of Adenosquamous Carcinoma of the Lung

Article Type
Changed
Mon, 06/26/2023 - 12:18
Display Headline
An Evolving Understanding of Adenosquamous Carcinoma of the Lung

Rajwanth Veluswamy, MD, MSCR
Adenosquamous carcinoma (ASC) of the lung is a rare, biphasic type of non-small cell lung cancer (NSCLC) that accounts for 2% to 4% of all lung cancers.1 According to the World Health Organization (WHO) classification, the composition of ASC includes both adenocarcinoma (AC) and squamous cell carcinoma (SCC) histologies, with each subtype comprising at least 10% of the tumor.2 As with other lung cancers, the average age at ASC diagnosis is about 70 years of age, it affects more men than women, and most patients are current or former smokers.3,4 Despite these similarities, mounting evidence suggests that the molecular and genomic features of ASC are unique and they remain poorly understood.5-8


Perhaps owing to the distinct genomics of these tumors, ASC of the lung is reported to be relatively aggressive compared to typical AC and SCC tumors. Studies indicate that ASCs at diagnosis have higher rates of lymph node invasion, metastasize rapidly, and carry a generally poor prognosis. Accordingly, the overall survival (OS) of patients with these tumors is relatively short compared to other NSCLC subtypes.2,3,8-10 In a 2022 population-based study of the SEER database, 5-year postsurgical survival rates for early-stage cancers were reportedly 65% for ASC vs 69% for SCC P=0.003 and 77% for AC P<0.001.While it is clear that underlying biology driving ASC differs from more typical NSCLC subtypes, there is a lack of effective treatment options specific to ASC and a paucity of clinical research available to support therapeutic decisions for patients with ASC histology. Current management of NSCLC is based primarily on the stage of the tumor, and clinical features of the patient. In a more personalized era of targeted treatments, tumor histology is used only to predict the presence of actionable mutations in adenocarcinomas.7,8 However, optimal treatment strategies for ASC remain a significant unmet need in lung cancer.


Diagnosis: Complex but Critically Important

Given the mixed histologies that characterize ASC of the lung, intratumoral heterogeneity often hinders and may delay diagnosis. Studies suggest that ASC is misdiagnosed as AC or SCC in at least half of biopsies prior to surgical pathology confirming an ASC diagnosis.11 In one retrospective study, nearly all ASC cases (98%) were either misdiagnosed or undiagnosed preoperatively.12 What’s more is that different types of biopsy samples may yield different results. One case report of a patient eventually diagnosed with ASC described 3 different results on workup: SCC on bronchial lavage and bronchial biopsy, AC on immunohistochemistry, and NSCLC undifferentiated on pleural effusion cytology.13 While a diagnosis can be made using biopsy and cytology samples, a definitive diagnosis may require larger samples (ie, several core biopsies or complete surgical resections) to fully evaluate all components of the tumor lesion.

Comprehensively evaluating entire tumor specimens can aid in further characterization ASC of the lung. ASCs may be sub-classified according to the proportions of AC and SCC histology components present. Tumors with either AC or SCC components comprising at least 60% of the tumor are referred to as AC- or SCC-predominant ASC, respectively. Those with a more even split of AC and SCC histologies (40% to 60% of each) are referred to as structure-balanced ASC and have been reported to have a better prognosis than either of the more imbalanced subtypes.9,14

Adding to the complexity of diagnosing ASC of the lung is its unclear histologic origin and the transitional nature of these tumors over time. Some studies have pointed to possible precursor lesions, including AC with squamous metaplasia, collision tumor, and high-grade mucoepidermoid tumors.15 Reports have also shown that the molecular and histological features of the primary tumor can differ from that of metastases/recurrences.16,17 In one case report, a patient with a resected ASC harboring an epidermal growth factor receptor (EGFR)-sensitizing mutation recurred several months later as SCC in the brain with the same EGFR mutation. A later recurrence in the lung was diagnosed as an AC and had the same EGFR mutation.16 In this example, if only the SCC component had been diagnosed, molecular testing would likely have never been ordered and the potentially actionable EGFR mutation would have been left undetected. Therefore, careful and accurate diagnosis of ASC is critically important in guiding testing for driver mutations, as well as in informing treatment choices in ASC.

Genomics

Studies indicate that ASC of the lung exhibits genomic features of both AC and SCC, with standard immunohistochemical profiles represented in each component. As expected, TTF1 positivity is common in the AC component while p63 and CK5/6 are expressed in the SCC component.18 However, evidence also indicates that ASC of the lung is a distinct entity rather than being a simple hybrid of AC and SCC histologies. That is, despite the seemingly dichotomous nature of ASC, this type of tumor is thought to have unique molecular and genomic features that have not yet been fully identified.5-8

While the genomics of AC and SCC of the lung have been well studied, the inherent intratumoral heterogeneity that defines ASC, together with its relative rarity, complicates its analysis. There is a paucity of data available, but several groups have conducted molecular testing to better understand the genotype of ASC and potentially discover predictors about prognosis and treatment. To date, most studies on ASC lung samples have been small, and while some groups have reported overlapping results, other findings contrast with one another. In one of the most recent and comprehensive studies published on the topic, Wang et al. used next-generation sequencing (NGS) to identify a wide range of somatic mutations in 124 Chinese patients with ASC of the lung, including TP53 (66.9%), CDKN2A (21%), TERT (21%), and LRP1B (18.5%).6 Importantly, they found high rates of EGFR mutations (54.8%), of which 45.6% were EGFR 19del, 38.2% were EGFR L858R and 29.4% were EGFR amplifications.

Notably, not all studies have found such a strikingly increased rate of EGFR mutations in ASC versus AC of the lung.19 Other actionable mutations were found in the analysis by Wang and colleagues, including ALK and ROS1 fusions. Regarding known predictors of immunogenicity in these tumors, a subset of patients were associated with high tumor mutational burden (TMB), which was correlated with mutations in ARID2, BRCA1, and KEAP1. Immunohistochemical analyses demonstrated half of patients were positive for PD-L1 (≥ 1% tumor proportion score [TPS]).6 Interestingly, another study showed that PD-L1 expression in ASC differed between SCC (30% to 40%) and AC (11% to 15%) components.20

Actionable mutation rates (ie, EGFR, ALK) in AC are known to vary between Asian and White patients, a finding that seems to be similar in ASC of the lung as well, although it is less clear given the limited sample size of ASC studies. Vassella et al. performed NGS and fluorescence in situ hybridization (FISH) on ASC samples from 16 White patients and found that 30% had EGFR mutations, while Tochigi et al reported an EGFR mutation rate of 13% in a study of 23 Western patients.5,12,21 In their analysis, Vassella and colleagues also found a high rate of mutations in the PI3K pathway (25%), but no KRAS mutations, which are the most common molecular driver in typical AC (30%), and thus supporting the notion that ASC has its own molecular genomic profile, distinct from AC or SCC.5,21 Also of interest in this study was the finding that classifier miR-205 expression was intermediate between that of classical AC and SCC, suggesting that ASC of the lung may alternatively represent a transitional stage between these tumor types rather than an unrelated entity.5 These findings, along with others that have been reported on the genomic landscape of ASC, have advanced our understanding of the underlying biology of this malignancy, but also highlight the unmet need for more research to improve our ability to personalize treatment for ASCs.

Treatment

Owing to the heterogeneity of ASC of the lung, as well as its complex and incompletely characterized genomic landscape, treating patients with these tumors is challenging. In general, stage-based treatment approaches are used to manage ASC. The current treatment paradigm of all NSCLC has dramatically changed in recent years, with increasing incorporation of targeted treatments and immunotherapies across all stages and histologic types. Considering ASCs are composed of glandular cell components, they can contain substantial levels of relevant actionable driver mutations as described above. Therefore, if ASC is diagnosed or if a SCC has a glandular component,molecular testing is recommended and supported by guidelines, even on surgical specimens where EGFR may be targeted as adjuvant treatment.23 However, while targeting actionable mutations and the PD1/PDL1 axis has been studied extensively in AC and SCC in all stages, the impact of these markers in ASC is unknown because patients with this histologic subtype are frequently excluded from clinical trials.

For patients with ASC and actionable mutations, EGFR inhibitors have been perhaps the best-studied targeted therapies. EGFR inhibitors have yielded responses in ASC, but the benefit has been highly variable in small case series and generally inferior to outcomes in patients with AC alone.19 Ongoing clinical trials are aiming to better understand the effects of EGFR inhibitors in ASC. As one example, first-line almonertinib is being compared to paclitaxel/carboplatin in the phase 2 ARISE clinical trial, which is specifically enrolling patients with EGFR mutation-positive locally advanced or metastatic pulmonary ASC (Clinicaltrials.gov NCT04354961). Most other reported studies are case studies or retrospective in nature.

Given that outcomes are usually reported from single patients or a group of only a few patients, contradictory findings are not uncommon. For example, crizotinib, a multi-kinase inhibitor approved for the treatment of advanced or metastatic ALK-positive and ROS1-positive NSCLC, was reported to have a clinical response in an ASC in a patient with recurrent ALK-positive disease which lasted for just over one year.24 However, the response to second-line crizotinib in a case report of female non-smoking patient with ROS1-positive ASC was only 4 months.25 Newer, more specific kinase inhibitors are currently in clinical practice and trials of ALK and ROS1 NSCLCs; however, their efficacy is ASC remains unclear.

In the absence of driver mutations, the optimal choice of chemotherapy (often given with immunotherapy) for neo-/adjuvant therapy or for metastatic disease has not yet been identified. While the AC component might typically be treated with pemetrexed plus a platinum agent, the SCC component may be better treated with taxane plus a platinum agent.23 Especially in cases where neither histologic subtype is predominant, it can be difficult to decide which combination may be suitable for an individual patient. Whether the relative proportion of AC and SCC components affect treatment outcomes is not yet known. Outcomes of pemetrexed-based chemotherapy have been reported in a case study of 2 patients with relapsed disease harboring ALK and ROS1 mutations, pemetrexed alone or as part of a combination regimen (with pembrolizumab and carboplatin) was able to maintain stable disease for at least a year.26

While immune checkpoint inhibitors, either as monotherapy or in combination with chemotherapy, are currently recommended for patients with NSCLC23, few studies have reported outcomes of patients with ASC specifically. One recent real-world analysis by Li et al. evaluated the effect of immunotherapy in 46 patients with ASC, of which 18 (39%) did not contain actionable driver mutations and 18 (39%) had unknown mutational status.27 In this study, 28% of the overall cohort responded to checkpoint inhibitors, the median progression-free survival was 6 months, and the median OS was 24.7 months. Notably, similar efficacy was observed in the 20 patients receiving immunotherapy monotherapy vs 26 patients who received combination immunotherapy plus chemotherapy.27 Among 4 patients with EGFR mutations, 2 received immunotherapy monotherapy and progressed immediately compared to the other 2 receiving combination immunotherapy with chemotherapy achieving disease control and improved OS (18 months).

As exemplified by the select few cases summarized above, conventional treatments used in NSCLC have achieved only modest responses in ASC, most with a shorter response duration. The lack of specific treatment strategies for ASC, based on our understanding of underlying tumor biology, limits optimal treatment outcomes for this increasingly common diagnosis. Novel therapies are sorely needed. A consensus should be developed to either study novel treatments specifically in this subtype or allow for the incorporation of ASCs into future NSCLC clinical trials.

Click to read more from 2023 Rare Diseases Report: Cancers

References
  1. Ruffini E, Rena O, Oliaro A, Filosso PL, Bongiovanni M, Arslanian A, Papalia E, Maggi G. Lung tumors with mixed histologic pattern. Clinico-pathologic characteristics and prognostic significance. Eur J Cardiothorac Surg. 2002; 22:701–707. doi:10.1016/s1010-7940(02)00481-5
  2. Almonertinib versus paclitaxel plus carboplatin as first-line treatment in patients with EGFR mutation positive locally advanced or metastatic pulmonary adenosquamous carcinoma (ARISE). ClinicalTrials.gov website. Accessed March 7, 2023. https://clinicaltrials.gov/ct2/show/NCT0435496
  3. Maeda H, Matsumura A, Kawabata T, et al. Adenosquamous carcinoma of the lung: surgical results as compared with squamous cell and adeno¬carcinoma cases. Eur J Cardiothorac Surg. 2012;41:357–361. doi:10.1016/j.ejcts.2011.05.050
  4. Wang T, Zhou J, Wang Y, et al. Clinicopathological characteristics and prognosis of resectable lung adenosquamous carcinoma: a population-based study of the SEER database. Jpn J Clin Oncol. 2022;52:1191-1200. doi:10.1093/jjco/hyac096
  5. Vassella E, Langsch S, Dettmer MS, et al. Molecular profiling of lung adenosquamous carcinoma: a hybrid or genuine type? Oncotarget. 2015;6:23905-23916. doi:10.18632/oncotarget.4163
  6. Wang H, Liu J, Zhu S, et al. Comprehensive analyses of genomic features and mutational signatures in adenosquamous carcinoma of the lung. Front Oncol. 2022;12:945843. doi:10.3389/fonc.2022.945843
  7. Li C, Lu H. Adenosquamous carcinoma of the lung. Onco Targets Ther. 2018;11:4829-4835. doi:10.2147/OTT.S164574
  8. Wang J, Wang Y, Tong M, Pan H, Li D. Research progress of the clinicopathologic features of lung adenosquamous carcinoma. Onco Targets Ther. 2018;11:7011-7017. doi:10.2147/OTT.S179904
  9. Gawrychowski J, Brulinski K, Malinowski E, Papla B. Prognosis and survival after radical resection of primary adenosquamous lung carcinoma. Eur J Cardiothorac Surg. 2005; 27:686–692. doi:10.1016/j.ejcts.2004.12.030
  10. Cooke DT, Nguyen DV, Yang Y, Chen SL, Yu C, Calhoun RF. Survival comparison of adenosquamous, squamous cell, and adenocarcinoma of the lung after lobectomy. Annal Thorac Surg. 2010; 90:943–948. doi:10.1016/j.athoracsur.2010.05.025
  11. Damadoglu E, Aybatli A, Yalçinsoy M, et al. Adenosquamous carcinoma of the lung (an analysis of 13 cases). Tuberk Toraks. 2005;53:161–166. https://pubmed.ncbi.nlm.nih.gov/16100653/
  12. Mordant P, Grand B, Cazes A, et al. Adenosquamous carcinoma of the lung: surgical management, pathologic characteristics, and prognostic implications. Ann Thorac Surg. 2013;95:1189–1195. doi:10.1016/j.athoracsur.2012.12.037
  13. Shelton DA, Rana DN, Holbrook M, Taylor P, Bailey S. Adenosquamous carcinoma of the lung diagnosed by cytology? A diagnostic dilemma. Diagn Cytopathol. 2012;40:830–833. doi:10.1002/dc.21664
  14. Zhao H, Yang H, Yao F, et al. Improved survival associated with a balanced structure between adenomatous and squamous components in patients with adenosquamous carcinoma of the lung. Eur J Surg Oncol. 2016;42:1699–1706. doi:10.1016/j.ejso.2016.05.009
  15. Shimizu J,Oda M,Hayashi Y,Nonomura A,Watanabe YA. Clinicopathological Study of resected cases of adenosquamous carcinoma of the lung. Chest. 1996; 109: 989-994. doi:10.1378/chest.109.4.989
  16. Burkart J, Shilo K, Zhao W, Ozkan E, Ajam A, Otterson GA. Metastatic squamous cell carcinoma component from an adenosquamous carcinoma of the lung with Identical epidermal growth factor receptor mutations. Case Rep Pulmonol. 2015;2015:283875. doi:10.1155/2015/283875
  17. Du C, Li Z, Wang Z, Wang L, Tian YU. Stereotactic aspiration combined with gamma knife radiosurgery for the treatment of cystic brainstem metastasis originating from lung adenosquamous carcinoma: A case report. Oncol Lett. 2015;9:1607–1613. doi:10.3892/ol.2015.2968
  18. Mukhopadhyay S, Katzenstein ALA. Subclassification of non-small cell lung carcinomas lacking morphologic differentiation on biopsy specimens: Utility of an immuno-histochemical panel containing TTF-1, napsin A, p63, and CK5/6. Am J Surg Pathol. 2011; 35:15–25. doi:10.1097/PAS.0b013e3182036d05
  19. Song X, Wang Z. Clinical efficacy evaluation of tyrosine kinase inhibitors for nonadenocarcinoma lung cancer patients harboring EGFR-sensitizing mutations. Onco Targets Ther. 2017;10:3119-3122. doi:10.2147/OTT.S134523
  20. Shi X, Wu S, Sun J, Liu Y, Zeng X, Liang Z. PD-L1 expression in lung adenosquamous carcinomas compared with the more common variants of non-small cell lung cancer. Sci Rep. 2017;7:46209. doi:10.1038/srep46209
  21. Cancer Genome Atlas Research N. Comprehensive molec¬ular profiling of lung adenocarcinoma. Nature. 2014; 511:543–550. doi:10.1038/nature13385
  22. Tochigi N, Dacic S, Nikiforova M, Cieply KM, Yousem SA. Adenosquamous carcinoma of the lung: a microdissection study of KRAS and EGFR mutational and amplification status in a western patient population. Am J Clin Pathol. 2011; 135:783–789. doi:10.1309/AJCP08IQZAOGYLFL
  23. National Comprehensive Cancer Network®. NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®). Non-small cell lung cancer. Version 2.2023. February 17, 2023. https://www.nccn.org/professionals/physician_gls/pdf/nscl.pdf. Accessed March 7, 2023.
  24. Chaft JE, Rekhtman N, Ladanyi M, Riely GJ. ALK-rearranged lung cancer: adenosquamous lung cancer masquerading as pure squamous carcinoma. J Thorac Oncol. 2012;7:768–769. doi:10.1097/JTO.0b013e31824c9485 
  25. Cheng Y, Yang J, Wang D, Yan D. ROS1 fusion lung adenosquamous carcinoma patient with short-term clinical benefit after crizotinib treatment: a case report. Ann Transl Med. 2022;10:157. doi:10.21037/atm-21-6754
  26. Patil J, Nie Y, Aisner DL, Camidge DR. Case report: significant clinical benefit from pemetrexed-based therapy in ROS-1 and ALK-rearranged lung cancer with adenosquamous histology. Front Oncol. 2022;11:788245. doi:10.3389/fonc.2021.788245
  27. Li C, Zheng X, Li P, et al. Heterogeneity of tumor immune microenvironment and real-world analysis of immunotherapy efficacy in lung adenosquamous carcinoma. Front Immunol. 2022;13:944812. doi:10.3389/fimmu.2022.944812

 

Author and Disclosure Information

Rajwanth Veluswamy, MD, MSCR
Assistant Professor of Medicine, Hematology and Medical Oncology
Icahn School of Medicine at Mount Sinai
New York, NY

Rajwanth R. Veluswamy, MD, MSCR, has disclosed the following relevant financial relationships:

Serve(d) as a director, officer, partner, employee, advisor, consultant, or trustee for: AstraZeneca; Boehringer Ingelheim; Merus; Novocure; Merck; Regeneron; Beigene; G1 Therapeutics; Novartis; BerGenBio.

Serve(d) as a speaker or a member of a speakers bureau for: AstraZeneca

Received research grant from: Bristol-Myers Squibb; Onconova; AstraZeneca; Boehringer Ingelheim

Publications
Topics
Author and Disclosure Information

Rajwanth Veluswamy, MD, MSCR
Assistant Professor of Medicine, Hematology and Medical Oncology
Icahn School of Medicine at Mount Sinai
New York, NY

Rajwanth R. Veluswamy, MD, MSCR, has disclosed the following relevant financial relationships:

Serve(d) as a director, officer, partner, employee, advisor, consultant, or trustee for: AstraZeneca; Boehringer Ingelheim; Merus; Novocure; Merck; Regeneron; Beigene; G1 Therapeutics; Novartis; BerGenBio.

Serve(d) as a speaker or a member of a speakers bureau for: AstraZeneca

Received research grant from: Bristol-Myers Squibb; Onconova; AstraZeneca; Boehringer Ingelheim

Author and Disclosure Information

Rajwanth Veluswamy, MD, MSCR
Assistant Professor of Medicine, Hematology and Medical Oncology
Icahn School of Medicine at Mount Sinai
New York, NY

Rajwanth R. Veluswamy, MD, MSCR, has disclosed the following relevant financial relationships:

Serve(d) as a director, officer, partner, employee, advisor, consultant, or trustee for: AstraZeneca; Boehringer Ingelheim; Merus; Novocure; Merck; Regeneron; Beigene; G1 Therapeutics; Novartis; BerGenBio.

Serve(d) as a speaker or a member of a speakers bureau for: AstraZeneca

Received research grant from: Bristol-Myers Squibb; Onconova; AstraZeneca; Boehringer Ingelheim

Rajwanth Veluswamy, MD, MSCR
Adenosquamous carcinoma (ASC) of the lung is a rare, biphasic type of non-small cell lung cancer (NSCLC) that accounts for 2% to 4% of all lung cancers.1 According to the World Health Organization (WHO) classification, the composition of ASC includes both adenocarcinoma (AC) and squamous cell carcinoma (SCC) histologies, with each subtype comprising at least 10% of the tumor.2 As with other lung cancers, the average age at ASC diagnosis is about 70 years of age, it affects more men than women, and most patients are current or former smokers.3,4 Despite these similarities, mounting evidence suggests that the molecular and genomic features of ASC are unique and they remain poorly understood.5-8


Perhaps owing to the distinct genomics of these tumors, ASC of the lung is reported to be relatively aggressive compared to typical AC and SCC tumors. Studies indicate that ASCs at diagnosis have higher rates of lymph node invasion, metastasize rapidly, and carry a generally poor prognosis. Accordingly, the overall survival (OS) of patients with these tumors is relatively short compared to other NSCLC subtypes.2,3,8-10 In a 2022 population-based study of the SEER database, 5-year postsurgical survival rates for early-stage cancers were reportedly 65% for ASC vs 69% for SCC P=0.003 and 77% for AC P<0.001.While it is clear that underlying biology driving ASC differs from more typical NSCLC subtypes, there is a lack of effective treatment options specific to ASC and a paucity of clinical research available to support therapeutic decisions for patients with ASC histology. Current management of NSCLC is based primarily on the stage of the tumor, and clinical features of the patient. In a more personalized era of targeted treatments, tumor histology is used only to predict the presence of actionable mutations in adenocarcinomas.7,8 However, optimal treatment strategies for ASC remain a significant unmet need in lung cancer.


Diagnosis: Complex but Critically Important

Given the mixed histologies that characterize ASC of the lung, intratumoral heterogeneity often hinders and may delay diagnosis. Studies suggest that ASC is misdiagnosed as AC or SCC in at least half of biopsies prior to surgical pathology confirming an ASC diagnosis.11 In one retrospective study, nearly all ASC cases (98%) were either misdiagnosed or undiagnosed preoperatively.12 What’s more is that different types of biopsy samples may yield different results. One case report of a patient eventually diagnosed with ASC described 3 different results on workup: SCC on bronchial lavage and bronchial biopsy, AC on immunohistochemistry, and NSCLC undifferentiated on pleural effusion cytology.13 While a diagnosis can be made using biopsy and cytology samples, a definitive diagnosis may require larger samples (ie, several core biopsies or complete surgical resections) to fully evaluate all components of the tumor lesion.

Comprehensively evaluating entire tumor specimens can aid in further characterization ASC of the lung. ASCs may be sub-classified according to the proportions of AC and SCC histology components present. Tumors with either AC or SCC components comprising at least 60% of the tumor are referred to as AC- or SCC-predominant ASC, respectively. Those with a more even split of AC and SCC histologies (40% to 60% of each) are referred to as structure-balanced ASC and have been reported to have a better prognosis than either of the more imbalanced subtypes.9,14

Adding to the complexity of diagnosing ASC of the lung is its unclear histologic origin and the transitional nature of these tumors over time. Some studies have pointed to possible precursor lesions, including AC with squamous metaplasia, collision tumor, and high-grade mucoepidermoid tumors.15 Reports have also shown that the molecular and histological features of the primary tumor can differ from that of metastases/recurrences.16,17 In one case report, a patient with a resected ASC harboring an epidermal growth factor receptor (EGFR)-sensitizing mutation recurred several months later as SCC in the brain with the same EGFR mutation. A later recurrence in the lung was diagnosed as an AC and had the same EGFR mutation.16 In this example, if only the SCC component had been diagnosed, molecular testing would likely have never been ordered and the potentially actionable EGFR mutation would have been left undetected. Therefore, careful and accurate diagnosis of ASC is critically important in guiding testing for driver mutations, as well as in informing treatment choices in ASC.

Genomics

Studies indicate that ASC of the lung exhibits genomic features of both AC and SCC, with standard immunohistochemical profiles represented in each component. As expected, TTF1 positivity is common in the AC component while p63 and CK5/6 are expressed in the SCC component.18 However, evidence also indicates that ASC of the lung is a distinct entity rather than being a simple hybrid of AC and SCC histologies. That is, despite the seemingly dichotomous nature of ASC, this type of tumor is thought to have unique molecular and genomic features that have not yet been fully identified.5-8

While the genomics of AC and SCC of the lung have been well studied, the inherent intratumoral heterogeneity that defines ASC, together with its relative rarity, complicates its analysis. There is a paucity of data available, but several groups have conducted molecular testing to better understand the genotype of ASC and potentially discover predictors about prognosis and treatment. To date, most studies on ASC lung samples have been small, and while some groups have reported overlapping results, other findings contrast with one another. In one of the most recent and comprehensive studies published on the topic, Wang et al. used next-generation sequencing (NGS) to identify a wide range of somatic mutations in 124 Chinese patients with ASC of the lung, including TP53 (66.9%), CDKN2A (21%), TERT (21%), and LRP1B (18.5%).6 Importantly, they found high rates of EGFR mutations (54.8%), of which 45.6% were EGFR 19del, 38.2% were EGFR L858R and 29.4% were EGFR amplifications.

Notably, not all studies have found such a strikingly increased rate of EGFR mutations in ASC versus AC of the lung.19 Other actionable mutations were found in the analysis by Wang and colleagues, including ALK and ROS1 fusions. Regarding known predictors of immunogenicity in these tumors, a subset of patients were associated with high tumor mutational burden (TMB), which was correlated with mutations in ARID2, BRCA1, and KEAP1. Immunohistochemical analyses demonstrated half of patients were positive for PD-L1 (≥ 1% tumor proportion score [TPS]).6 Interestingly, another study showed that PD-L1 expression in ASC differed between SCC (30% to 40%) and AC (11% to 15%) components.20

Actionable mutation rates (ie, EGFR, ALK) in AC are known to vary between Asian and White patients, a finding that seems to be similar in ASC of the lung as well, although it is less clear given the limited sample size of ASC studies. Vassella et al. performed NGS and fluorescence in situ hybridization (FISH) on ASC samples from 16 White patients and found that 30% had EGFR mutations, while Tochigi et al reported an EGFR mutation rate of 13% in a study of 23 Western patients.5,12,21 In their analysis, Vassella and colleagues also found a high rate of mutations in the PI3K pathway (25%), but no KRAS mutations, which are the most common molecular driver in typical AC (30%), and thus supporting the notion that ASC has its own molecular genomic profile, distinct from AC or SCC.5,21 Also of interest in this study was the finding that classifier miR-205 expression was intermediate between that of classical AC and SCC, suggesting that ASC of the lung may alternatively represent a transitional stage between these tumor types rather than an unrelated entity.5 These findings, along with others that have been reported on the genomic landscape of ASC, have advanced our understanding of the underlying biology of this malignancy, but also highlight the unmet need for more research to improve our ability to personalize treatment for ASCs.

Treatment

Owing to the heterogeneity of ASC of the lung, as well as its complex and incompletely characterized genomic landscape, treating patients with these tumors is challenging. In general, stage-based treatment approaches are used to manage ASC. The current treatment paradigm of all NSCLC has dramatically changed in recent years, with increasing incorporation of targeted treatments and immunotherapies across all stages and histologic types. Considering ASCs are composed of glandular cell components, they can contain substantial levels of relevant actionable driver mutations as described above. Therefore, if ASC is diagnosed or if a SCC has a glandular component,molecular testing is recommended and supported by guidelines, even on surgical specimens where EGFR may be targeted as adjuvant treatment.23 However, while targeting actionable mutations and the PD1/PDL1 axis has been studied extensively in AC and SCC in all stages, the impact of these markers in ASC is unknown because patients with this histologic subtype are frequently excluded from clinical trials.

For patients with ASC and actionable mutations, EGFR inhibitors have been perhaps the best-studied targeted therapies. EGFR inhibitors have yielded responses in ASC, but the benefit has been highly variable in small case series and generally inferior to outcomes in patients with AC alone.19 Ongoing clinical trials are aiming to better understand the effects of EGFR inhibitors in ASC. As one example, first-line almonertinib is being compared to paclitaxel/carboplatin in the phase 2 ARISE clinical trial, which is specifically enrolling patients with EGFR mutation-positive locally advanced or metastatic pulmonary ASC (Clinicaltrials.gov NCT04354961). Most other reported studies are case studies or retrospective in nature.

Given that outcomes are usually reported from single patients or a group of only a few patients, contradictory findings are not uncommon. For example, crizotinib, a multi-kinase inhibitor approved for the treatment of advanced or metastatic ALK-positive and ROS1-positive NSCLC, was reported to have a clinical response in an ASC in a patient with recurrent ALK-positive disease which lasted for just over one year.24 However, the response to second-line crizotinib in a case report of female non-smoking patient with ROS1-positive ASC was only 4 months.25 Newer, more specific kinase inhibitors are currently in clinical practice and trials of ALK and ROS1 NSCLCs; however, their efficacy is ASC remains unclear.

In the absence of driver mutations, the optimal choice of chemotherapy (often given with immunotherapy) for neo-/adjuvant therapy or for metastatic disease has not yet been identified. While the AC component might typically be treated with pemetrexed plus a platinum agent, the SCC component may be better treated with taxane plus a platinum agent.23 Especially in cases where neither histologic subtype is predominant, it can be difficult to decide which combination may be suitable for an individual patient. Whether the relative proportion of AC and SCC components affect treatment outcomes is not yet known. Outcomes of pemetrexed-based chemotherapy have been reported in a case study of 2 patients with relapsed disease harboring ALK and ROS1 mutations, pemetrexed alone or as part of a combination regimen (with pembrolizumab and carboplatin) was able to maintain stable disease for at least a year.26

While immune checkpoint inhibitors, either as monotherapy or in combination with chemotherapy, are currently recommended for patients with NSCLC23, few studies have reported outcomes of patients with ASC specifically. One recent real-world analysis by Li et al. evaluated the effect of immunotherapy in 46 patients with ASC, of which 18 (39%) did not contain actionable driver mutations and 18 (39%) had unknown mutational status.27 In this study, 28% of the overall cohort responded to checkpoint inhibitors, the median progression-free survival was 6 months, and the median OS was 24.7 months. Notably, similar efficacy was observed in the 20 patients receiving immunotherapy monotherapy vs 26 patients who received combination immunotherapy plus chemotherapy.27 Among 4 patients with EGFR mutations, 2 received immunotherapy monotherapy and progressed immediately compared to the other 2 receiving combination immunotherapy with chemotherapy achieving disease control and improved OS (18 months).

As exemplified by the select few cases summarized above, conventional treatments used in NSCLC have achieved only modest responses in ASC, most with a shorter response duration. The lack of specific treatment strategies for ASC, based on our understanding of underlying tumor biology, limits optimal treatment outcomes for this increasingly common diagnosis. Novel therapies are sorely needed. A consensus should be developed to either study novel treatments specifically in this subtype or allow for the incorporation of ASCs into future NSCLC clinical trials.

Click to read more from 2023 Rare Diseases Report: Cancers

Rajwanth Veluswamy, MD, MSCR
Adenosquamous carcinoma (ASC) of the lung is a rare, biphasic type of non-small cell lung cancer (NSCLC) that accounts for 2% to 4% of all lung cancers.1 According to the World Health Organization (WHO) classification, the composition of ASC includes both adenocarcinoma (AC) and squamous cell carcinoma (SCC) histologies, with each subtype comprising at least 10% of the tumor.2 As with other lung cancers, the average age at ASC diagnosis is about 70 years of age, it affects more men than women, and most patients are current or former smokers.3,4 Despite these similarities, mounting evidence suggests that the molecular and genomic features of ASC are unique and they remain poorly understood.5-8


Perhaps owing to the distinct genomics of these tumors, ASC of the lung is reported to be relatively aggressive compared to typical AC and SCC tumors. Studies indicate that ASCs at diagnosis have higher rates of lymph node invasion, metastasize rapidly, and carry a generally poor prognosis. Accordingly, the overall survival (OS) of patients with these tumors is relatively short compared to other NSCLC subtypes.2,3,8-10 In a 2022 population-based study of the SEER database, 5-year postsurgical survival rates for early-stage cancers were reportedly 65% for ASC vs 69% for SCC P=0.003 and 77% for AC P<0.001.While it is clear that underlying biology driving ASC differs from more typical NSCLC subtypes, there is a lack of effective treatment options specific to ASC and a paucity of clinical research available to support therapeutic decisions for patients with ASC histology. Current management of NSCLC is based primarily on the stage of the tumor, and clinical features of the patient. In a more personalized era of targeted treatments, tumor histology is used only to predict the presence of actionable mutations in adenocarcinomas.7,8 However, optimal treatment strategies for ASC remain a significant unmet need in lung cancer.


Diagnosis: Complex but Critically Important

Given the mixed histologies that characterize ASC of the lung, intratumoral heterogeneity often hinders and may delay diagnosis. Studies suggest that ASC is misdiagnosed as AC or SCC in at least half of biopsies prior to surgical pathology confirming an ASC diagnosis.11 In one retrospective study, nearly all ASC cases (98%) were either misdiagnosed or undiagnosed preoperatively.12 What’s more is that different types of biopsy samples may yield different results. One case report of a patient eventually diagnosed with ASC described 3 different results on workup: SCC on bronchial lavage and bronchial biopsy, AC on immunohistochemistry, and NSCLC undifferentiated on pleural effusion cytology.13 While a diagnosis can be made using biopsy and cytology samples, a definitive diagnosis may require larger samples (ie, several core biopsies or complete surgical resections) to fully evaluate all components of the tumor lesion.

Comprehensively evaluating entire tumor specimens can aid in further characterization ASC of the lung. ASCs may be sub-classified according to the proportions of AC and SCC histology components present. Tumors with either AC or SCC components comprising at least 60% of the tumor are referred to as AC- or SCC-predominant ASC, respectively. Those with a more even split of AC and SCC histologies (40% to 60% of each) are referred to as structure-balanced ASC and have been reported to have a better prognosis than either of the more imbalanced subtypes.9,14

Adding to the complexity of diagnosing ASC of the lung is its unclear histologic origin and the transitional nature of these tumors over time. Some studies have pointed to possible precursor lesions, including AC with squamous metaplasia, collision tumor, and high-grade mucoepidermoid tumors.15 Reports have also shown that the molecular and histological features of the primary tumor can differ from that of metastases/recurrences.16,17 In one case report, a patient with a resected ASC harboring an epidermal growth factor receptor (EGFR)-sensitizing mutation recurred several months later as SCC in the brain with the same EGFR mutation. A later recurrence in the lung was diagnosed as an AC and had the same EGFR mutation.16 In this example, if only the SCC component had been diagnosed, molecular testing would likely have never been ordered and the potentially actionable EGFR mutation would have been left undetected. Therefore, careful and accurate diagnosis of ASC is critically important in guiding testing for driver mutations, as well as in informing treatment choices in ASC.

Genomics

Studies indicate that ASC of the lung exhibits genomic features of both AC and SCC, with standard immunohistochemical profiles represented in each component. As expected, TTF1 positivity is common in the AC component while p63 and CK5/6 are expressed in the SCC component.18 However, evidence also indicates that ASC of the lung is a distinct entity rather than being a simple hybrid of AC and SCC histologies. That is, despite the seemingly dichotomous nature of ASC, this type of tumor is thought to have unique molecular and genomic features that have not yet been fully identified.5-8

While the genomics of AC and SCC of the lung have been well studied, the inherent intratumoral heterogeneity that defines ASC, together with its relative rarity, complicates its analysis. There is a paucity of data available, but several groups have conducted molecular testing to better understand the genotype of ASC and potentially discover predictors about prognosis and treatment. To date, most studies on ASC lung samples have been small, and while some groups have reported overlapping results, other findings contrast with one another. In one of the most recent and comprehensive studies published on the topic, Wang et al. used next-generation sequencing (NGS) to identify a wide range of somatic mutations in 124 Chinese patients with ASC of the lung, including TP53 (66.9%), CDKN2A (21%), TERT (21%), and LRP1B (18.5%).6 Importantly, they found high rates of EGFR mutations (54.8%), of which 45.6% were EGFR 19del, 38.2% were EGFR L858R and 29.4% were EGFR amplifications.

Notably, not all studies have found such a strikingly increased rate of EGFR mutations in ASC versus AC of the lung.19 Other actionable mutations were found in the analysis by Wang and colleagues, including ALK and ROS1 fusions. Regarding known predictors of immunogenicity in these tumors, a subset of patients were associated with high tumor mutational burden (TMB), which was correlated with mutations in ARID2, BRCA1, and KEAP1. Immunohistochemical analyses demonstrated half of patients were positive for PD-L1 (≥ 1% tumor proportion score [TPS]).6 Interestingly, another study showed that PD-L1 expression in ASC differed between SCC (30% to 40%) and AC (11% to 15%) components.20

Actionable mutation rates (ie, EGFR, ALK) in AC are known to vary between Asian and White patients, a finding that seems to be similar in ASC of the lung as well, although it is less clear given the limited sample size of ASC studies. Vassella et al. performed NGS and fluorescence in situ hybridization (FISH) on ASC samples from 16 White patients and found that 30% had EGFR mutations, while Tochigi et al reported an EGFR mutation rate of 13% in a study of 23 Western patients.5,12,21 In their analysis, Vassella and colleagues also found a high rate of mutations in the PI3K pathway (25%), but no KRAS mutations, which are the most common molecular driver in typical AC (30%), and thus supporting the notion that ASC has its own molecular genomic profile, distinct from AC or SCC.5,21 Also of interest in this study was the finding that classifier miR-205 expression was intermediate between that of classical AC and SCC, suggesting that ASC of the lung may alternatively represent a transitional stage between these tumor types rather than an unrelated entity.5 These findings, along with others that have been reported on the genomic landscape of ASC, have advanced our understanding of the underlying biology of this malignancy, but also highlight the unmet need for more research to improve our ability to personalize treatment for ASCs.

Treatment

Owing to the heterogeneity of ASC of the lung, as well as its complex and incompletely characterized genomic landscape, treating patients with these tumors is challenging. In general, stage-based treatment approaches are used to manage ASC. The current treatment paradigm of all NSCLC has dramatically changed in recent years, with increasing incorporation of targeted treatments and immunotherapies across all stages and histologic types. Considering ASCs are composed of glandular cell components, they can contain substantial levels of relevant actionable driver mutations as described above. Therefore, if ASC is diagnosed or if a SCC has a glandular component,molecular testing is recommended and supported by guidelines, even on surgical specimens where EGFR may be targeted as adjuvant treatment.23 However, while targeting actionable mutations and the PD1/PDL1 axis has been studied extensively in AC and SCC in all stages, the impact of these markers in ASC is unknown because patients with this histologic subtype are frequently excluded from clinical trials.

For patients with ASC and actionable mutations, EGFR inhibitors have been perhaps the best-studied targeted therapies. EGFR inhibitors have yielded responses in ASC, but the benefit has been highly variable in small case series and generally inferior to outcomes in patients with AC alone.19 Ongoing clinical trials are aiming to better understand the effects of EGFR inhibitors in ASC. As one example, first-line almonertinib is being compared to paclitaxel/carboplatin in the phase 2 ARISE clinical trial, which is specifically enrolling patients with EGFR mutation-positive locally advanced or metastatic pulmonary ASC (Clinicaltrials.gov NCT04354961). Most other reported studies are case studies or retrospective in nature.

Given that outcomes are usually reported from single patients or a group of only a few patients, contradictory findings are not uncommon. For example, crizotinib, a multi-kinase inhibitor approved for the treatment of advanced or metastatic ALK-positive and ROS1-positive NSCLC, was reported to have a clinical response in an ASC in a patient with recurrent ALK-positive disease which lasted for just over one year.24 However, the response to second-line crizotinib in a case report of female non-smoking patient with ROS1-positive ASC was only 4 months.25 Newer, more specific kinase inhibitors are currently in clinical practice and trials of ALK and ROS1 NSCLCs; however, their efficacy is ASC remains unclear.

In the absence of driver mutations, the optimal choice of chemotherapy (often given with immunotherapy) for neo-/adjuvant therapy or for metastatic disease has not yet been identified. While the AC component might typically be treated with pemetrexed plus a platinum agent, the SCC component may be better treated with taxane plus a platinum agent.23 Especially in cases where neither histologic subtype is predominant, it can be difficult to decide which combination may be suitable for an individual patient. Whether the relative proportion of AC and SCC components affect treatment outcomes is not yet known. Outcomes of pemetrexed-based chemotherapy have been reported in a case study of 2 patients with relapsed disease harboring ALK and ROS1 mutations, pemetrexed alone or as part of a combination regimen (with pembrolizumab and carboplatin) was able to maintain stable disease for at least a year.26

While immune checkpoint inhibitors, either as monotherapy or in combination with chemotherapy, are currently recommended for patients with NSCLC23, few studies have reported outcomes of patients with ASC specifically. One recent real-world analysis by Li et al. evaluated the effect of immunotherapy in 46 patients with ASC, of which 18 (39%) did not contain actionable driver mutations and 18 (39%) had unknown mutational status.27 In this study, 28% of the overall cohort responded to checkpoint inhibitors, the median progression-free survival was 6 months, and the median OS was 24.7 months. Notably, similar efficacy was observed in the 20 patients receiving immunotherapy monotherapy vs 26 patients who received combination immunotherapy plus chemotherapy.27 Among 4 patients with EGFR mutations, 2 received immunotherapy monotherapy and progressed immediately compared to the other 2 receiving combination immunotherapy with chemotherapy achieving disease control and improved OS (18 months).

As exemplified by the select few cases summarized above, conventional treatments used in NSCLC have achieved only modest responses in ASC, most with a shorter response duration. The lack of specific treatment strategies for ASC, based on our understanding of underlying tumor biology, limits optimal treatment outcomes for this increasingly common diagnosis. Novel therapies are sorely needed. A consensus should be developed to either study novel treatments specifically in this subtype or allow for the incorporation of ASCs into future NSCLC clinical trials.

Click to read more from 2023 Rare Diseases Report: Cancers

References
  1. Ruffini E, Rena O, Oliaro A, Filosso PL, Bongiovanni M, Arslanian A, Papalia E, Maggi G. Lung tumors with mixed histologic pattern. Clinico-pathologic characteristics and prognostic significance. Eur J Cardiothorac Surg. 2002; 22:701–707. doi:10.1016/s1010-7940(02)00481-5
  2. Almonertinib versus paclitaxel plus carboplatin as first-line treatment in patients with EGFR mutation positive locally advanced or metastatic pulmonary adenosquamous carcinoma (ARISE). ClinicalTrials.gov website. Accessed March 7, 2023. https://clinicaltrials.gov/ct2/show/NCT0435496
  3. Maeda H, Matsumura A, Kawabata T, et al. Adenosquamous carcinoma of the lung: surgical results as compared with squamous cell and adeno¬carcinoma cases. Eur J Cardiothorac Surg. 2012;41:357–361. doi:10.1016/j.ejcts.2011.05.050
  4. Wang T, Zhou J, Wang Y, et al. Clinicopathological characteristics and prognosis of resectable lung adenosquamous carcinoma: a population-based study of the SEER database. Jpn J Clin Oncol. 2022;52:1191-1200. doi:10.1093/jjco/hyac096
  5. Vassella E, Langsch S, Dettmer MS, et al. Molecular profiling of lung adenosquamous carcinoma: a hybrid or genuine type? Oncotarget. 2015;6:23905-23916. doi:10.18632/oncotarget.4163
  6. Wang H, Liu J, Zhu S, et al. Comprehensive analyses of genomic features and mutational signatures in adenosquamous carcinoma of the lung. Front Oncol. 2022;12:945843. doi:10.3389/fonc.2022.945843
  7. Li C, Lu H. Adenosquamous carcinoma of the lung. Onco Targets Ther. 2018;11:4829-4835. doi:10.2147/OTT.S164574
  8. Wang J, Wang Y, Tong M, Pan H, Li D. Research progress of the clinicopathologic features of lung adenosquamous carcinoma. Onco Targets Ther. 2018;11:7011-7017. doi:10.2147/OTT.S179904
  9. Gawrychowski J, Brulinski K, Malinowski E, Papla B. Prognosis and survival after radical resection of primary adenosquamous lung carcinoma. Eur J Cardiothorac Surg. 2005; 27:686–692. doi:10.1016/j.ejcts.2004.12.030
  10. Cooke DT, Nguyen DV, Yang Y, Chen SL, Yu C, Calhoun RF. Survival comparison of adenosquamous, squamous cell, and adenocarcinoma of the lung after lobectomy. Annal Thorac Surg. 2010; 90:943–948. doi:10.1016/j.athoracsur.2010.05.025
  11. Damadoglu E, Aybatli A, Yalçinsoy M, et al. Adenosquamous carcinoma of the lung (an analysis of 13 cases). Tuberk Toraks. 2005;53:161–166. https://pubmed.ncbi.nlm.nih.gov/16100653/
  12. Mordant P, Grand B, Cazes A, et al. Adenosquamous carcinoma of the lung: surgical management, pathologic characteristics, and prognostic implications. Ann Thorac Surg. 2013;95:1189–1195. doi:10.1016/j.athoracsur.2012.12.037
  13. Shelton DA, Rana DN, Holbrook M, Taylor P, Bailey S. Adenosquamous carcinoma of the lung diagnosed by cytology? A diagnostic dilemma. Diagn Cytopathol. 2012;40:830–833. doi:10.1002/dc.21664
  14. Zhao H, Yang H, Yao F, et al. Improved survival associated with a balanced structure between adenomatous and squamous components in patients with adenosquamous carcinoma of the lung. Eur J Surg Oncol. 2016;42:1699–1706. doi:10.1016/j.ejso.2016.05.009
  15. Shimizu J,Oda M,Hayashi Y,Nonomura A,Watanabe YA. Clinicopathological Study of resected cases of adenosquamous carcinoma of the lung. Chest. 1996; 109: 989-994. doi:10.1378/chest.109.4.989
  16. Burkart J, Shilo K, Zhao W, Ozkan E, Ajam A, Otterson GA. Metastatic squamous cell carcinoma component from an adenosquamous carcinoma of the lung with Identical epidermal growth factor receptor mutations. Case Rep Pulmonol. 2015;2015:283875. doi:10.1155/2015/283875
  17. Du C, Li Z, Wang Z, Wang L, Tian YU. Stereotactic aspiration combined with gamma knife radiosurgery for the treatment of cystic brainstem metastasis originating from lung adenosquamous carcinoma: A case report. Oncol Lett. 2015;9:1607–1613. doi:10.3892/ol.2015.2968
  18. Mukhopadhyay S, Katzenstein ALA. Subclassification of non-small cell lung carcinomas lacking morphologic differentiation on biopsy specimens: Utility of an immuno-histochemical panel containing TTF-1, napsin A, p63, and CK5/6. Am J Surg Pathol. 2011; 35:15–25. doi:10.1097/PAS.0b013e3182036d05
  19. Song X, Wang Z. Clinical efficacy evaluation of tyrosine kinase inhibitors for nonadenocarcinoma lung cancer patients harboring EGFR-sensitizing mutations. Onco Targets Ther. 2017;10:3119-3122. doi:10.2147/OTT.S134523
  20. Shi X, Wu S, Sun J, Liu Y, Zeng X, Liang Z. PD-L1 expression in lung adenosquamous carcinomas compared with the more common variants of non-small cell lung cancer. Sci Rep. 2017;7:46209. doi:10.1038/srep46209
  21. Cancer Genome Atlas Research N. Comprehensive molec¬ular profiling of lung adenocarcinoma. Nature. 2014; 511:543–550. doi:10.1038/nature13385
  22. Tochigi N, Dacic S, Nikiforova M, Cieply KM, Yousem SA. Adenosquamous carcinoma of the lung: a microdissection study of KRAS and EGFR mutational and amplification status in a western patient population. Am J Clin Pathol. 2011; 135:783–789. doi:10.1309/AJCP08IQZAOGYLFL
  23. National Comprehensive Cancer Network®. NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®). Non-small cell lung cancer. Version 2.2023. February 17, 2023. https://www.nccn.org/professionals/physician_gls/pdf/nscl.pdf. Accessed March 7, 2023.
  24. Chaft JE, Rekhtman N, Ladanyi M, Riely GJ. ALK-rearranged lung cancer: adenosquamous lung cancer masquerading as pure squamous carcinoma. J Thorac Oncol. 2012;7:768–769. doi:10.1097/JTO.0b013e31824c9485 
  25. Cheng Y, Yang J, Wang D, Yan D. ROS1 fusion lung adenosquamous carcinoma patient with short-term clinical benefit after crizotinib treatment: a case report. Ann Transl Med. 2022;10:157. doi:10.21037/atm-21-6754
  26. Patil J, Nie Y, Aisner DL, Camidge DR. Case report: significant clinical benefit from pemetrexed-based therapy in ROS-1 and ALK-rearranged lung cancer with adenosquamous histology. Front Oncol. 2022;11:788245. doi:10.3389/fonc.2021.788245
  27. Li C, Zheng X, Li P, et al. Heterogeneity of tumor immune microenvironment and real-world analysis of immunotherapy efficacy in lung adenosquamous carcinoma. Front Immunol. 2022;13:944812. doi:10.3389/fimmu.2022.944812

 

References
  1. Ruffini E, Rena O, Oliaro A, Filosso PL, Bongiovanni M, Arslanian A, Papalia E, Maggi G. Lung tumors with mixed histologic pattern. Clinico-pathologic characteristics and prognostic significance. Eur J Cardiothorac Surg. 2002; 22:701–707. doi:10.1016/s1010-7940(02)00481-5
  2. Almonertinib versus paclitaxel plus carboplatin as first-line treatment in patients with EGFR mutation positive locally advanced or metastatic pulmonary adenosquamous carcinoma (ARISE). ClinicalTrials.gov website. Accessed March 7, 2023. https://clinicaltrials.gov/ct2/show/NCT0435496
  3. Maeda H, Matsumura A, Kawabata T, et al. Adenosquamous carcinoma of the lung: surgical results as compared with squamous cell and adeno¬carcinoma cases. Eur J Cardiothorac Surg. 2012;41:357–361. doi:10.1016/j.ejcts.2011.05.050
  4. Wang T, Zhou J, Wang Y, et al. Clinicopathological characteristics and prognosis of resectable lung adenosquamous carcinoma: a population-based study of the SEER database. Jpn J Clin Oncol. 2022;52:1191-1200. doi:10.1093/jjco/hyac096
  5. Vassella E, Langsch S, Dettmer MS, et al. Molecular profiling of lung adenosquamous carcinoma: a hybrid or genuine type? Oncotarget. 2015;6:23905-23916. doi:10.18632/oncotarget.4163
  6. Wang H, Liu J, Zhu S, et al. Comprehensive analyses of genomic features and mutational signatures in adenosquamous carcinoma of the lung. Front Oncol. 2022;12:945843. doi:10.3389/fonc.2022.945843
  7. Li C, Lu H. Adenosquamous carcinoma of the lung. Onco Targets Ther. 2018;11:4829-4835. doi:10.2147/OTT.S164574
  8. Wang J, Wang Y, Tong M, Pan H, Li D. Research progress of the clinicopathologic features of lung adenosquamous carcinoma. Onco Targets Ther. 2018;11:7011-7017. doi:10.2147/OTT.S179904
  9. Gawrychowski J, Brulinski K, Malinowski E, Papla B. Prognosis and survival after radical resection of primary adenosquamous lung carcinoma. Eur J Cardiothorac Surg. 2005; 27:686–692. doi:10.1016/j.ejcts.2004.12.030
  10. Cooke DT, Nguyen DV, Yang Y, Chen SL, Yu C, Calhoun RF. Survival comparison of adenosquamous, squamous cell, and adenocarcinoma of the lung after lobectomy. Annal Thorac Surg. 2010; 90:943–948. doi:10.1016/j.athoracsur.2010.05.025
  11. Damadoglu E, Aybatli A, Yalçinsoy M, et al. Adenosquamous carcinoma of the lung (an analysis of 13 cases). Tuberk Toraks. 2005;53:161–166. https://pubmed.ncbi.nlm.nih.gov/16100653/
  12. Mordant P, Grand B, Cazes A, et al. Adenosquamous carcinoma of the lung: surgical management, pathologic characteristics, and prognostic implications. Ann Thorac Surg. 2013;95:1189–1195. doi:10.1016/j.athoracsur.2012.12.037
  13. Shelton DA, Rana DN, Holbrook M, Taylor P, Bailey S. Adenosquamous carcinoma of the lung diagnosed by cytology? A diagnostic dilemma. Diagn Cytopathol. 2012;40:830–833. doi:10.1002/dc.21664
  14. Zhao H, Yang H, Yao F, et al. Improved survival associated with a balanced structure between adenomatous and squamous components in patients with adenosquamous carcinoma of the lung. Eur J Surg Oncol. 2016;42:1699–1706. doi:10.1016/j.ejso.2016.05.009
  15. Shimizu J,Oda M,Hayashi Y,Nonomura A,Watanabe YA. Clinicopathological Study of resected cases of adenosquamous carcinoma of the lung. Chest. 1996; 109: 989-994. doi:10.1378/chest.109.4.989
  16. Burkart J, Shilo K, Zhao W, Ozkan E, Ajam A, Otterson GA. Metastatic squamous cell carcinoma component from an adenosquamous carcinoma of the lung with Identical epidermal growth factor receptor mutations. Case Rep Pulmonol. 2015;2015:283875. doi:10.1155/2015/283875
  17. Du C, Li Z, Wang Z, Wang L, Tian YU. Stereotactic aspiration combined with gamma knife radiosurgery for the treatment of cystic brainstem metastasis originating from lung adenosquamous carcinoma: A case report. Oncol Lett. 2015;9:1607–1613. doi:10.3892/ol.2015.2968
  18. Mukhopadhyay S, Katzenstein ALA. Subclassification of non-small cell lung carcinomas lacking morphologic differentiation on biopsy specimens: Utility of an immuno-histochemical panel containing TTF-1, napsin A, p63, and CK5/6. Am J Surg Pathol. 2011; 35:15–25. doi:10.1097/PAS.0b013e3182036d05
  19. Song X, Wang Z. Clinical efficacy evaluation of tyrosine kinase inhibitors for nonadenocarcinoma lung cancer patients harboring EGFR-sensitizing mutations. Onco Targets Ther. 2017;10:3119-3122. doi:10.2147/OTT.S134523
  20. Shi X, Wu S, Sun J, Liu Y, Zeng X, Liang Z. PD-L1 expression in lung adenosquamous carcinomas compared with the more common variants of non-small cell lung cancer. Sci Rep. 2017;7:46209. doi:10.1038/srep46209
  21. Cancer Genome Atlas Research N. Comprehensive molec¬ular profiling of lung adenocarcinoma. Nature. 2014; 511:543–550. doi:10.1038/nature13385
  22. Tochigi N, Dacic S, Nikiforova M, Cieply KM, Yousem SA. Adenosquamous carcinoma of the lung: a microdissection study of KRAS and EGFR mutational and amplification status in a western patient population. Am J Clin Pathol. 2011; 135:783–789. doi:10.1309/AJCP08IQZAOGYLFL
  23. National Comprehensive Cancer Network®. NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®). Non-small cell lung cancer. Version 2.2023. February 17, 2023. https://www.nccn.org/professionals/physician_gls/pdf/nscl.pdf. Accessed March 7, 2023.
  24. Chaft JE, Rekhtman N, Ladanyi M, Riely GJ. ALK-rearranged lung cancer: adenosquamous lung cancer masquerading as pure squamous carcinoma. J Thorac Oncol. 2012;7:768–769. doi:10.1097/JTO.0b013e31824c9485 
  25. Cheng Y, Yang J, Wang D, Yan D. ROS1 fusion lung adenosquamous carcinoma patient with short-term clinical benefit after crizotinib treatment: a case report. Ann Transl Med. 2022;10:157. doi:10.21037/atm-21-6754
  26. Patil J, Nie Y, Aisner DL, Camidge DR. Case report: significant clinical benefit from pemetrexed-based therapy in ROS-1 and ALK-rearranged lung cancer with adenosquamous histology. Front Oncol. 2022;11:788245. doi:10.3389/fonc.2021.788245
  27. Li C, Zheng X, Li P, et al. Heterogeneity of tumor immune microenvironment and real-world analysis of immunotherapy efficacy in lung adenosquamous carcinoma. Front Immunol. 2022;13:944812. doi:10.3389/fimmu.2022.944812

 

Publications
Publications
Topics
Article Type
Display Headline
An Evolving Understanding of Adenosquamous Carcinoma of the Lung
Display Headline
An Evolving Understanding of Adenosquamous Carcinoma of the Lung
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Wed, 06/07/2023 - 17:00
Un-Gate On Date
Wed, 06/07/2023 - 17:00
Use ProPublica
CFC Schedule Remove Status
Wed, 06/07/2023 - 17:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Pharmacotherapy underprescribed for alcohol use disorder

Article Type
Changed
Tue, 06/27/2023 - 09:26

Health care providers are missing opportunities to give medical treatment to high-risk individuals hospitalized for alcohol use disorder (AUD), a national analysis of Medicare beneficiaries reported.

Increasing such patients’ access to psychiatric care and addiction medicine, as well as encouraging medication prescribing by generalists and nonaddiction specialists, are remedial strategies recommended by lead author Eden Y. Bernstein, MD, of the division of general internal medicine at Massachusetts General Hospital and Harvard Medical School, both in Boston, and colleagues.

“Hospitalizations for alcohol use disorder are common,” Dr. Bernstein said in an interview. “Our work shows they represent an underutilized opportunity to engage patients with appropriate treatment, including initiation of medications for alcohol use disorder.”

There is a pressing need for such treatment strategies since 29 million U.S. adults have AUD, and alcohol contributes to more than 140,000 deaths annually, the authors noted.

Rarely initiated either at hospital discharge or during follow-up care, medical therapy for AUD was more likely to be provided to younger patients and those involved with psychiatric care or addiction medicine, Dr. Bernstein’s group reported in Annals of Internal Medicine.Hospital admissions, they argued, give patients more access to clinicians and social workers and the vulnerability experienced during hospitalization may motivate behavioral change.


 

National study

The cohort included 28,601 AUD hospitalizations for 20,401 unique Medicare patients from 2015 to 2017. About 30% of admissions were for women and about 72% for non-Hispanic Blacks. Discharge initiation of medication for AUD was defined as a pharmacy claim for naltrexone, acamprosate, or disulfiram from the day before discharge to 2 days after.

Overall, just 206 patients (0.7%) initiated medication for AUD within 2 days of discharge and 364 (1.3%) started it within 30 days. Among those discharged with a primary diagnosis of AUD, only 70 (2.3%) started medical therapy within 2 days.

The most predictive demographic factor for discharge medication for AUD was younger age: 18-39 years versus 75 years and older (adjusted odds ratio, 3.87; 95% confidence interval, 1.34-11.16).

Initiation of medication for AUD should involve a long-term treatment plan, according to Dr. Bernstein’s group, and if that is not feasible during hospitalization, patients should be referred for outpatient treatment.

An accompanying editorial agrees that the results offer strong evidence of a missed opportunity to address AUD at a potential flexion point. “Hospitalization is a critical touch point for identifying and treating AUD,” wrote Michael F. Mayo-Smith, MD, MPH, of White River Junction (Vt.) VA Medical Center, and Geisel School of Medicine at Dartmouth, Hanover, N.H., and David Lawrence, MD, of the VA Greater Los Angeles Healthcare System and the University of California, Los Angeles.

An intentional discharge protocol can be effective, they noted, as evidenced by a 2014 report in which this approach increased medication-assisted treatment from 0% to 64% in tandem with a decrease in all-cause, 30-day readmission rates.

“There is also growing interest in inpatient addiction consultation services, which have shown [medication] for AUD treatment initiation rates of up to 70% as well as improved engagement in posthospital treatment,” Dr. Mayo-Smith and Dr. Lawrence wrote.

Minority populations need particular attention, they added. “Unfortunately, the availability of evidence-based treatments for AUD does not by itself lead to improved care. We need strategies for widespread adoption so that patients can realize the benefits of these treatments.”

Dr. Bernstein reported funding support from a National Research Service Award and the Massachusetts General Hospital division of general internal medicine; he disclosed fees from Alosa Health. One coauthor was supported by the Agency for Healthcare Research and Quality. Another was supported by the National Institute on Aging and reported relationships with the American College of Cardiology, Boston OIAC Pepper Center, American Heart Association, and US Deprescribing Research Network. Dr. Mayo-Smith disclosed no competing interests. Dr. Lawrence reported fees related to presentations at DDW 2023 and the California Society of Addiction Medicine 2022.

Publications
Topics
Sections

Health care providers are missing opportunities to give medical treatment to high-risk individuals hospitalized for alcohol use disorder (AUD), a national analysis of Medicare beneficiaries reported.

Increasing such patients’ access to psychiatric care and addiction medicine, as well as encouraging medication prescribing by generalists and nonaddiction specialists, are remedial strategies recommended by lead author Eden Y. Bernstein, MD, of the division of general internal medicine at Massachusetts General Hospital and Harvard Medical School, both in Boston, and colleagues.

“Hospitalizations for alcohol use disorder are common,” Dr. Bernstein said in an interview. “Our work shows they represent an underutilized opportunity to engage patients with appropriate treatment, including initiation of medications for alcohol use disorder.”

There is a pressing need for such treatment strategies since 29 million U.S. adults have AUD, and alcohol contributes to more than 140,000 deaths annually, the authors noted.

Rarely initiated either at hospital discharge or during follow-up care, medical therapy for AUD was more likely to be provided to younger patients and those involved with psychiatric care or addiction medicine, Dr. Bernstein’s group reported in Annals of Internal Medicine.Hospital admissions, they argued, give patients more access to clinicians and social workers and the vulnerability experienced during hospitalization may motivate behavioral change.


 

National study

The cohort included 28,601 AUD hospitalizations for 20,401 unique Medicare patients from 2015 to 2017. About 30% of admissions were for women and about 72% for non-Hispanic Blacks. Discharge initiation of medication for AUD was defined as a pharmacy claim for naltrexone, acamprosate, or disulfiram from the day before discharge to 2 days after.

Overall, just 206 patients (0.7%) initiated medication for AUD within 2 days of discharge and 364 (1.3%) started it within 30 days. Among those discharged with a primary diagnosis of AUD, only 70 (2.3%) started medical therapy within 2 days.

The most predictive demographic factor for discharge medication for AUD was younger age: 18-39 years versus 75 years and older (adjusted odds ratio, 3.87; 95% confidence interval, 1.34-11.16).

Initiation of medication for AUD should involve a long-term treatment plan, according to Dr. Bernstein’s group, and if that is not feasible during hospitalization, patients should be referred for outpatient treatment.

An accompanying editorial agrees that the results offer strong evidence of a missed opportunity to address AUD at a potential flexion point. “Hospitalization is a critical touch point for identifying and treating AUD,” wrote Michael F. Mayo-Smith, MD, MPH, of White River Junction (Vt.) VA Medical Center, and Geisel School of Medicine at Dartmouth, Hanover, N.H., and David Lawrence, MD, of the VA Greater Los Angeles Healthcare System and the University of California, Los Angeles.

An intentional discharge protocol can be effective, they noted, as evidenced by a 2014 report in which this approach increased medication-assisted treatment from 0% to 64% in tandem with a decrease in all-cause, 30-day readmission rates.

“There is also growing interest in inpatient addiction consultation services, which have shown [medication] for AUD treatment initiation rates of up to 70% as well as improved engagement in posthospital treatment,” Dr. Mayo-Smith and Dr. Lawrence wrote.

Minority populations need particular attention, they added. “Unfortunately, the availability of evidence-based treatments for AUD does not by itself lead to improved care. We need strategies for widespread adoption so that patients can realize the benefits of these treatments.”

Dr. Bernstein reported funding support from a National Research Service Award and the Massachusetts General Hospital division of general internal medicine; he disclosed fees from Alosa Health. One coauthor was supported by the Agency for Healthcare Research and Quality. Another was supported by the National Institute on Aging and reported relationships with the American College of Cardiology, Boston OIAC Pepper Center, American Heart Association, and US Deprescribing Research Network. Dr. Mayo-Smith disclosed no competing interests. Dr. Lawrence reported fees related to presentations at DDW 2023 and the California Society of Addiction Medicine 2022.

Health care providers are missing opportunities to give medical treatment to high-risk individuals hospitalized for alcohol use disorder (AUD), a national analysis of Medicare beneficiaries reported.

Increasing such patients’ access to psychiatric care and addiction medicine, as well as encouraging medication prescribing by generalists and nonaddiction specialists, are remedial strategies recommended by lead author Eden Y. Bernstein, MD, of the division of general internal medicine at Massachusetts General Hospital and Harvard Medical School, both in Boston, and colleagues.

“Hospitalizations for alcohol use disorder are common,” Dr. Bernstein said in an interview. “Our work shows they represent an underutilized opportunity to engage patients with appropriate treatment, including initiation of medications for alcohol use disorder.”

There is a pressing need for such treatment strategies since 29 million U.S. adults have AUD, and alcohol contributes to more than 140,000 deaths annually, the authors noted.

Rarely initiated either at hospital discharge or during follow-up care, medical therapy for AUD was more likely to be provided to younger patients and those involved with psychiatric care or addiction medicine, Dr. Bernstein’s group reported in Annals of Internal Medicine.Hospital admissions, they argued, give patients more access to clinicians and social workers and the vulnerability experienced during hospitalization may motivate behavioral change.


 

National study

The cohort included 28,601 AUD hospitalizations for 20,401 unique Medicare patients from 2015 to 2017. About 30% of admissions were for women and about 72% for non-Hispanic Blacks. Discharge initiation of medication for AUD was defined as a pharmacy claim for naltrexone, acamprosate, or disulfiram from the day before discharge to 2 days after.

Overall, just 206 patients (0.7%) initiated medication for AUD within 2 days of discharge and 364 (1.3%) started it within 30 days. Among those discharged with a primary diagnosis of AUD, only 70 (2.3%) started medical therapy within 2 days.

The most predictive demographic factor for discharge medication for AUD was younger age: 18-39 years versus 75 years and older (adjusted odds ratio, 3.87; 95% confidence interval, 1.34-11.16).

Initiation of medication for AUD should involve a long-term treatment plan, according to Dr. Bernstein’s group, and if that is not feasible during hospitalization, patients should be referred for outpatient treatment.

An accompanying editorial agrees that the results offer strong evidence of a missed opportunity to address AUD at a potential flexion point. “Hospitalization is a critical touch point for identifying and treating AUD,” wrote Michael F. Mayo-Smith, MD, MPH, of White River Junction (Vt.) VA Medical Center, and Geisel School of Medicine at Dartmouth, Hanover, N.H., and David Lawrence, MD, of the VA Greater Los Angeles Healthcare System and the University of California, Los Angeles.

An intentional discharge protocol can be effective, they noted, as evidenced by a 2014 report in which this approach increased medication-assisted treatment from 0% to 64% in tandem with a decrease in all-cause, 30-day readmission rates.

“There is also growing interest in inpatient addiction consultation services, which have shown [medication] for AUD treatment initiation rates of up to 70% as well as improved engagement in posthospital treatment,” Dr. Mayo-Smith and Dr. Lawrence wrote.

Minority populations need particular attention, they added. “Unfortunately, the availability of evidence-based treatments for AUD does not by itself lead to improved care. We need strategies for widespread adoption so that patients can realize the benefits of these treatments.”

Dr. Bernstein reported funding support from a National Research Service Award and the Massachusetts General Hospital division of general internal medicine; he disclosed fees from Alosa Health. One coauthor was supported by the Agency for Healthcare Research and Quality. Another was supported by the National Institute on Aging and reported relationships with the American College of Cardiology, Boston OIAC Pepper Center, American Heart Association, and US Deprescribing Research Network. Dr. Mayo-Smith disclosed no competing interests. Dr. Lawrence reported fees related to presentations at DDW 2023 and the California Society of Addiction Medicine 2022.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM ANNALS OF INTERNAL MEDICINE

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Progress in Ovarian Cancer: Discovery of Fallopian Tube Involvement

Article Type
Changed
Mon, 06/26/2023 - 12:16
Display Headline
Progress in Ovarian Cancer: Discovery of Fallopian Tube Involvement

Ronny Drapkin, MD, PhD
The field of ovarian cancer has experienced a paradigm shift; ovarian cancer is now known to most often arise from the fallopian tubes.1 The ovaries can act as a magnet for tumor cells that may originate elsewhere in the body. Moreover, it has been found that relatively simple risk-reducing interventions may virtually eliminate progression to invasive disease in the ovaries.1 These types of discoveries—and others—are igniting new research into novel approaches to improving outcomes for patients with ovarian cancer.

 

Incidence and Mortality

By 2040, the number of women diagnosed with ovarian cancer annually worldwide is expected to increase by 100% in low Human Development Index (HDI) countries, and by 19-28% in high HDI countries.2 The causes of this increasing incidence are likely to be multifactorial, including both hereditary and modifiable risk factors.3 In addition to increasing population size, the growing prevalence of obesity, estrogen exposures, and nulliparity are particularly pertinent as potential causes of the rising incidence of ovarian cancer in younger women. The number of ovarian cancer-related deaths is also projected to rise from about 200,000 to nearly 314,000 annually, an increase of over 50% from 2020.2,4 Although outcomes in developed regions and nations continue to improve somewhat, 5-year survival rates range from 36% to 46%.5 These outcomes are nevertheless dismal when compared with 5-year survival rates from other cancer types, such as breast cancer, which are approaching 90%.6

Principal Histotypes

The principal histotypes in ovarian cancer are epithelial in origin and include high-grade serous carcinoma, clear-cell carcinoma, endometrioid carcinoma, low-grade serous carcinoma, and mucinous carcinoma. Other rarer types are nonepithelial, ie, arising from stromal or germ cell lines.7 Incidence rates appear to be affected over time by trends such as birth rates, use of combination oral contraceptives, and menopausal hormone therapy.8 Figure 1 shows that most ovarian cancers—approximately 70%—are high-grade serous carcinoma, although in Asian countries clear cell and endometrioid carcinomas comprise a higher proportion.9

Figure 1. Major Histotypes in Ovarian Cancer
Most ovarian cancers are epithelial carcinomas. High-grade serous carcinoma is the most common, whereas the other subtypes represent 10% or fewer cases each.9

Into the Fallopian Tube

One of the most salient and dramatic discoveries of the last 2 decades has been the finding that high-grade, clear-cell, and endometrioid tumors appear to arise from tissues not normally present in the ovary.1 As a result of risk-reducing efforts to prevent serous cancers in women with genetic predisposition to develop ovarian cancer (ie, those with BRCA1 or BRCA2 mutations), it became increasingly clear that many early cancers arose in the fallopian tube,10-12 with the distal portion—the fimbria—as the most common site of origin.13-16

Figure 2 depicts the female reproductive tract, including the location of the fimbria compared with the ovaries. Moreover, lesions observed in the fallopian tube fimbria—serous tubal intraepithelial carcinomas (STICs)—were identified as precursors of ovarian cancer, with a window of 7 years between development of STIC and the beginning of an ovarian cancer.14,16

 

Figure 2. Serous Tubal Intraepithelial Carcinomas
Lesions that develop in the fallopian tube fimbria, called serous tubal intraepithelial carcinomas (STICs), have been identified as precursors of ovarian cancer.

Early Detection

Early, localized ovarian cancer is asymptomatic; by the time a patient presents with symptoms, even with nonspecific abdominal complaints, the disease is almost invariably advanced. The concept of early detection has improved both the rate of cancer diagnoses and outcomes for some malignancies, such as cervical, colorectal, breast, and lung cancers,17 but this strategy is yet to be effectively applied in ovarian cancer. A large, population-based study, for instance, yielded negative results when multimodal screening (using both measurement of CA125 blood levels and transvaginal ultrasound imaging) failed to improve survival, even though such screening was able to detect lower stage disease.18 Emerging technologies, such as liquid biopsies and uterine lavage, which seek to detect potential biomarkers (new types of blood tests) of ovarian cancer at an early stage and closer to the site of tumor origin, are being investigated and refined but are not yet ready for clinical use, particularly at the population level for screening.19

Risk-Reducing Interventions

Use of oral contraceptives has been associated with a significant reduction in risk for ovarian cancer, but the potential risks (eg, increased risk for breast cancer, increased risk for venous thromboembolism) preclude its universal recommendation.20-22 Simple removal of the fallopian tube, salpingectomy, was proposed as a potential intervention to “intercept” the progression of a STIC to cancer. Researchers recently compared simple salpingectomy with salpingo-oophorectomy as a risk-reduction procedure in carriers of BRCA 1/2 pathogenic variants after they had completed childbearing.23

These investigators proposed that later removal of the ovaries would delay menopause and would contribute to fewer/less severe symptoms, such as hot flashes, disturbed sleep, and sexual issues, as well as maintain or improve overall quality of life. The hypothesis was supported by results, which showed that patients had better menopause-related quality of life after salpingectomy than after salpingo-oophorectomy, regardless of the use of hormone replacement therapy.23 The oncologic safety of this approach was subsequently demonstrated by other studies that showed a significantly lower incidence of ovarian cancers in women who had undergone opportunistic salpingectomy.22,24,25

An international prospective trial, TUBA-WISPII, is now underway to test the hypothesis that postponement of oophorectomy after salpingectomy is non-inferior to standard salpingo-oophorectomy in terms of ovarian cancer risk for patients at high risk.26

Treatment

First-Line Therapy
Currently, there are no durable curative therapies for ovarian cancer once advanced disease has been diagnosed.

Surgery plus platinum-based chemotherapy. Most patients, even those diagnosed with advanced disease, are treated initially with debulking surgery, ideally by a gynecologic oncologist, and adjuvant chemotherapy. Most ovarian carcinomas are initially platinum-sensitive, but resistance and disease recurrence are almost inevitable. According to the National Comprehensive Cancer Network (NCCN) guidelines for ovarian cancer,27 preferred chemotherapy regimens include paclitaxel and carboplatin with or without bevacizumab, docetaxel and carboplatin, or carboplatin and liposomal doxorubicin. Numerous other regimens, combinations, and agents are included in the guidelines to help providers customize treatment plans.

Neoadjuvant vs adjuvant regimens. Neoadjuvant chemotherapy has been used for other malignancies to gauge sensitivity to systemic treatments and to improve surgical margins.28 Thus far, though, outcomes in ovarian cancer have been similar whether patients were given neoadjuvant or adjuvant treatment in the perioperative period. Individualizing these decisions based on ability to surgically resect, patient age, tumor histology, disease stage, and performance status is recommended.29

Intraperitoneal chemotherapy. Other approaches have been explored to reduce risk for micrometastases after surgery. Hyperthermic intraperitoneal chemotherapy,32 for instance, administered immediately after cytoreductive surgery was studied as a technique that might prevent some of the risks and adverse effects associated with intraperitoneal chemotherapy.31 Results showed some improvement in progression-free survival and overall survival in a subgroup of patients who underwent interval cytoreductive surgery after neoadjuvant therapy, but no differences were observed for the larger population with advanced epithelial ovarian cancer. Adverse reactions to intraperitoneal chemotherapy were also observed.

Angiogenesis inhibition. Tumors need energy and oxygen to grow. Angiogenesis is the process of new blood vessel formation that provides the tumor with nutrients. Blocking angiogenesis can thwart tumor growth and improve patient outcomes. Bevacizumab is an antiangiogenic agent that has been extensively studied for 2 decades for many cancers including ovarian carcinoma. The NCCN guidelines note that bevacizumab may be considered as part of a first-line regimen with platinum agents, as maintenance in patients with wild-type or unknown BRCA mutation status and a good response to first-line therapy, or in combination with a poly (ADP-ribose) polymerase (PARP) inhibitor in eligible patients.27

PARP inhibitors. Approximately half of all high-grade serous ovarian carcinomas exhibit some defect in the ability to repair DNA damage using the homologous recombination (HR) pathway. These tumors include those with mutations in the BRCA1, BRCA2, and other HR genes. Defects in HR make tumors more dependent on back-up DNA repair systems, including the activity of PARP. PARP inhibitors were developed to specifically target HR-deficient tumors. To date, 3 PARP inhibitors have been approved for use in ovarian cancer—olaparib, rucaparib, and niraparib. Their use has expanded from later-line use in patients with BRCA1/2-mutated tumors to include frontline maintenance regimens for women with high-grade serous and high-grade endometrioid carcinomas, as well as women with recurrent disease.32 Numerous clinical trials are ongoing to develop next-generation PARP inhibitors and to explore their efficacy in combination with chemotherapy and other targeted agents.

Resistance and Disease Progression: Second-Line and Subsequent Treatment

A number of second-line and subsequent systemic treatment regimens may be considered when primary platinum-based chemotherapy and/or maintenance are no longer effective.33,34 As emphasized by the NCCN, a clinical trial is always an appropriate option, depending on eligibility, and sometimes a second cytoreductive surgery35,36 may be considered for patients who experience radiographic and/or clinical relapse after a long disease-free interval (6+ months). Each line of treatment is associated with progressively lower response rates and shorter durations of response. According to the NCCN guidelines, as patient performance status decreases and the toxicities of each line of therapy accumulate, assessment for palliative care should be considered and discussed.27

Investigational Approaches

With the high mortality rate associated with ovarian cancer, the challenges of detecting the disease at its early stages, and the lack of therapies that can significantly extend progression-free and overall survival in patients with advanced disease, many investigators are focused on novel treatment approaches. Preclinical observations, for instance, showing synergy between ataxia telangiectasia and RAD3-related (ATR) kinase inhibitors and PARP inhibitors led researchers to initiate a phase 2 study of olaparib plus ceralasertib (an ATR inhibitor) in patients with recurrent, platinum-resistant epithelial ovarian cancer.37 No objective responses were noted, but some signals of activity were seen among patients with BRCA1 mutations.

Due to success in other malignancies, immunotherapy is also being explored. Although some promising signals were reported at 6 months when nivolumab, a PD-1 (programmed cell death protein 1) inhibitor, and ipilimumab, a cytotoxic T-lymphocyte-associated antigen 4 antibody, were combined to treat patients with platinum-resistant epithelial ovarian cancer; final results are not yet available.38

Ovarian cancer is sometimes characterized as immunologically “cold.” This description means that immune cells, especially T cells, are not able to enter the tumor and destroy the cancer cells. It also means that these tumors are not as responsive to immune-based treatments. Therefore, some researchers are examining novel alternative immunotherapy strategies, such as chimeric antigen receptor T-cell (CAR-T) therapy.39 When a CAR T-cell encounters a tumor antigen, the CAR T-cell becomes activated. Activated CAR T-cells multiply, signal to other immune cells, and ultimately kill the tumor cells. Although CAR T-cell therapy has been tremendously successful in hematologic malignancies, to date, the benefits in solid tumors have been modest.39 However, there is significant enthusiasm for novel tumor antigens that can be targeted by CAR-T therapy, including mesothelin, folate receptor, Claudin-6, B7-H3, B7-H4, HER2, CD47, and L1-CAM, among others.40

Other investigational strategies include a p53 vaccine that would enhance the patient’s immunologic response to abnormal proteins produced by a mutated p53 gene, which is the most common finding in ovarian tumors.

Although researchers are investigating many approaches to treating advanced ovarian cancer, one strategy that has been pursued in other cancer settings—development of antibody-drug conjugates (ADCs)41—has seen promising results. In the late fall of 2022, the US Food and Drug Administration granted accelerated approval for mirvetuximab soravtansine-gynx for use in patients with a specific type of type of tumor (folate receptor alpha [FRα]-positive) when platinum resistance emerges.42 A companion diagnostic assay was also approved for selecting patients with FRα-positive disease. Several other clinical trials are investigating the efficacy of targeting other ovarian tumor antigens using the ADC approach. These targets include NaPi2b, mesothelin, B7-H4, Claudin-6, and Trop-2.43,44

Progress to Come 

Progress in ovarian cancer will be made through a multipronged approach that includes interventions that may proactively “intercept” the development of cancer (eg, salpingectomy for women planning to have other simple gynecologic procedures after childbearing is complete). Although prophylactic surgeries are often undertaken by individuals at high risk for ovarian cancer because of genetic findings, such as BRCA1/2 abnormalities, even women with normal risk may consider when planning tubal ligation, removing their tubes, and other routine procedures. A substantial number of malignant tumors, and associated morbidity and mortality, may be thwarted as a result. The question of whether to treat when a STIC is detected remains to be answered.

The search for better methods of early detection continues, as local therapies for early-stage disease are invariably more effective than treatments in the advanced and/or metastatic setting.

Finally, as with certain other malignancies, even in the advanced setting, effective, often targeted, treatments can significantly prolong both progression-free and overall survival, transforming an often-lethal disease into a chronic one that allows patients to enjoy a better life expectancy with good quality of life.

Click to read more from 2023 Rare Diseases Report: Cancers

References
  1. Karnezis AN, Cho KR, Gilks CB, Pearce CL, Huntsman DG. The disparate origins of ovarian cancers: pathogenesis and prevention strategies. Nat Rev Cancer. 2017;17(1):65-74. doi:10.1038/nrc.2016.113
  2. Cabasag CJ, Fagan PJ, Ferlay J, et al. Ovarian cancer today and tomorrow: A global assessment by world region and Human Development Index using GLOBOCAN 2020. Int J Cancer. 2022;151(9):1535-1541. doi:10.1002/ijc.34002
  3. Huang J, Chan WC, Ngai CH, et al. Worldwide burden, risk factors, and temporal trends of ovarian cancer: a global study. Cancers (Basel). 2022;14(9):2230. doi:10.3390/cancers14092230
  4. Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA Cancer J Clin. 2023;73(1):17-48. doi:10.3322/caac.21763
  5. World Ovarian Cancer Coalition. Ovarian cancer key stats. Accessed May 8, 2023. https://worldovariancancercoalition.org/about-ovarian-cancer/key-stats/
  6. American Cancer Society. Survival rates for breast cancer. Updated March 1, 2023. Accessed May 8, 2023. https://www.cancer.org/cancer/breast-cancer/understanding-a-breast-cancer-diagnosis/breast-cancer-survival-rates.html
  7. Reid BM, Permuth JB, Sellers TA. Epidemiology of ovarian cancer: a review. Cancer Biol Med. 2017;14(1):9-32. doi:10.20892/j.issn.2095-3941.2016.0084
  8. Phung MT, Pearce CL, Meza R, Jeon J. Trends of ovarian cancer incidence by histotype and race/ethnicity in the United States 1992–2019. Cancer Res Commun. 2023;3(1):1-8. doi:10.1158/2767-9764.CRC-22-0410
  9. Coburn SB, Bray F, Sherman ME, Trabert B. International patterns and trends in ovarian cancer incidence, overall and by histologic subtype. Int J Cancer. 2017;140(11):2451-2460. doi:10.1002/ijc.30676
  10. Kroeger PT Jr, Drapkin R. Pathogenesis and heterogeneity of ovarian cancer. Curr Obstet Gynecol. 2017;29(1):26-34. doi:10.1097/GCO.0000000000000340
  11. Shih lM, Wang Y, Wang TL. The origin of ovarian cancer species and precancerous landscape. Am J Pathol. 2021;191(1):26-39. doi:10.1016/j.ajpath.2020.09.006
  12. Meserve EEK, Brouwer J, Crum CP. Serous tubal intraepithelial neoplasia: the concept and its application. Mod Pathol. 2017;30(5):710-721. doi:10.1038/modpathol.2016.23
  13. Crum CP, Drapkin R, Kindelberger D, Medeiros F, Miron A, Lee Y. Lessons from BRCA: the tubal fimbria emerges as an origin for pelvic serous cancer. Clin Med Res. 2007;5(1):35-44. doi:10.3121/cmr.2007.702
  14. Wu RC, Wang P, Lin SF, et al. Genomic landscape and evolutionary trajectories of ovarian cancer precursor lesions. J Pathol. 2019;248(1):41-50. doi:10.1002/path.5219
  15. Eckert MA, Pan S, Hernandez KM, et al. Genomics of ovarian cancer progression reveals diverse metastatic trajectories including intraepithelial metastasis to the fallopian tube. Cancer Discov. 2016;6(12):1342-1351. doi:10.1158/2159-8290.CD-16-0607
  16. Labidi-Galy SI, Papp E, Hallberg D, et al. High grade serous ovarian carcinomas originate in the fallopian tube. Nat Comm. 2017;8(1):1093. doi:10.1038/s41467-017-00962-1
  17. Centers for Disease Control and Prevention; National Comprehensive Cancer Control Program (NCCP). Promoting early detection and treatment of cancer. Reviewed July 30, 2021. Accessed May 8, 2023. https://www.cdc.gov/cancer/ncccp/priorities/early-detection-treatment.htm
  18. Menon U, Gentry-Maharaj A, Burnell M, et al. Ovarian cancer population screening and mortality after long-term follow-up in the UK Collaborative Trial of Ovarian Cancer Screening (UKCTOCS): a randomised controlled trial. Lancet. 2021;397(10290):2182-2193. doi:10.1016/S0140-6736(21)00731-5
  19. Žilovic D, Ciurliene R, Sabaliauskaite R, Jarmalaitė S. Future screening prospects for ovarian cancer. Cancers (Basel). 2021;13(15):3840. doi:10.3390/cancers1315384
  20. Michels KA, Pfeiffer RM, Brinton LA, Trabert B. Modification of the associations between duration of oral contraceptive use and ovarian, endometrial, breast, and colorectal cancers. JAMA Oncol. 2018;4(4):516-521. doi:10.1001/jamaoncol.2017.4942
  21. Havrilesky LJ, Moorman PG, Lowery WJ, et al. Oral contraceptive pills as primary prevention for ovarian cancer: a systematic review and meta-analysis. Obstet Gynecol. 2013;122(1):139-147. doi:10.1097/AOG.0b013e318291c235
  22. Kotsopoulos J, Narod SA. Prophylactic salpingectomy for the prevention of ovarian cancer: who should we target? Int J Cancer. 2020;147(5):1245-1251. doi:10.1002/ijc.32916 
  23. Steenbeek MP, Harmsen MG, Hoogerbrugge N, et al. Association of salpingectomy with delayed oophorectomy versus salpingo-oophorectomy with quality of life in BRCA 1/2 pathogenic variant carriers. A nonrandomized controlled trial. JAMA Oncol. 2021;7(8):1203-1212. doi:10.1001/jamaoncol.2021.1590
  24. Hanley GE, Pearce CL, Talhouk A, et al. Outcomes from opportunistic salpingectomy for ovarian cancer prevention. JAMA Netw Open. 2022;5(2):e2147343. doi:10.1001/jamanetworkopen.2021.47343
  25. Society of Gynecologic Oncology (SGO). SGO clinical practice statement: salpingectomy for ovarian cancer prevention (SGO, November 2013). Published November 1, 2013. Accessed May 8, 2023. https://www.sgo.org/resources/sgo-clinical-practice-statement-salpingectomy-for-ovarian-cancer-prevention/
  26. Steenbeek MP, van Bommel MHD, intHout J, et al. TUBectomy with delayed oophorectomy as an alternative to risk-reducing salpingo-oophorectomy in high-risk women to assess the safety of prevention: the TUBA-WISP II study protocol [published online ahead of print, 2023 Apr 12]. Int J Gynecol Cancer. 2023;ijgc-2023-004377. doi:10.1136/ijgc-2023-004377
  27. National Comprehensive Cancer Network. NCCN clinical practice guidelines in oncology: Ovarian cancer including fallopian tube cancer and primary peritoneal cancer. Version 1.2023. December 22, 2022. Accessed May 8, 2023. https://www.nccn.org/professionals/physician_gls/pdf/ovarian.pdf
  28. Chawla A, Hunt KK, Mittendorf EA. Surgical considerations in patients receiving neoadjuvant systemic therapy. Future Oncol. 2012;8(3):239-250. doi:10.2217/fon.12.12
  29. Coleridge SL, Bryant A, Kehoe S, Morrison J. Neoadjuvant chemotherapy before surgery versus surgery followed by chemotherapy for initial treatment in advanced ovarian epithelial cancer. Cochrane Database Syst Rev. 2021;7(7):CD005343. doi:10.1002/14651858.CD005343.pub6
  30. Lim MC, Chang SJ, Park B, et al; HIPEC for Ovarian Cancer Collaborators. Survival after hyperthermic intraperitoneal chemotherapy and primary or interval cytoreductive surgery in ovarian cancer: a randomized clinical trial. JAMA Surg. 2022;157(5):374-383. doi:10.1001/jamasurg.2022.0143
  31. Walker JL, Brady MF, Wenzel L, et al. Randomized trial of intravenous versus intraperitoneal chemotherapy plus bevacizumab in advanced ovarian carcinoma: an NRG Oncology/Gynecologic Oncology Group study. J Clin Oncol. 2019;37(16):1380-1390. doi:10.1200/JCO.18.01568 
  32. Konstantinopoulos PA, Lheureux S, Moore KN. PARP inhibitors for ovarian cancer: current indications, future combinations, and novel assets in development to target DNA damage repair. Am Soc Clin Oncol Educ Book. 2020;40:1-16. doi:10.1200/EDBK_288015
  33. Markman M, Bookman MA. Second-line treatment of ovarian cancer. Oncologist. 2000;5(1):26-35. doi:10.1634/theoncologist.5-1-26
  34. Markman M. Pharmaceutical management of ovarian cancer: current status. Drugs. 2019;79(11):1231-1239. doi:10.1007/s40265-019-01158-1
  35. Bristow RE, Puri I, Chi DS. Cytoreductive surgery for recurrent ovarian cancer: a meta-analysis. Gynecol Oncol. 2009;112(1):265-274. doi:10.1016/j.ygyno.2008.08.033
  36. de Bree E, Michelakis D, Anagnostopoulou E. The current role of secondary cytoreductive surgery for recurrent ovarian cancer. Front Oncol. 2022;12:1029976. doi:10.3389/fonc.2022.1029976
  37. Shah PD, Wethington SL, Pagan C, et al. Combination ATR and PARP inhibitor (CAPRI): a phase 2 study of ceralasertib plus olaparib in patients with recurrent, platinum-resistant epithelial ovarian cancer. Gynecol Oncol. 2021;163(2): 246-253. doi:10.1016/j.ygyno.2021.08.024
  38. Borella F, Ghisoni E, Giannone G, et al. Immune checkpoint inhibitors in epithelial ovarian cancer: an overview on efficacy and future perspectives. Diagnostics (Basel). 2020;10(3):146. doi:10.3390/diagnostics10030146
  39. Wu JWY, Dand S, Doig L, et al. T-cell receptor therapy in the treatment of ovarian cancer: a mini review. Front Immunol. 2021;12:672502. doi:10.3389/fimmu.2021.672502
  40. Benard E, Casey NP, Inderberg EM, Wälchli S. SJI 2020 special issue: a catalogue of ovarian cancer targets for CAR therapy. Scand J Immunol. 2020;92(4):e12917 doi:10.1111/sji.12917
  41. Martín-Sabroso C, Lozza I, Torres-Suárez AI, Fraguas-Sánchez AI. Antibodyantineoplastic conjugates in gynecological malignancies: current status and future perspectives. Pharmaceutics. 2021;13(10):1705. doi:10.3390/pharmaceutics13101705
  42. US Food and Drug Administration. FDA grants accelerated approval to mirvetuximab soravtansine-gynx for FRα positive, platinum-resistant epithelial ovarian, fallopian tube, or peritoneal cancer. Published November 14, 2022. Accessed May 8, 2023. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-mirvetuximab-soravtansine-gynx-fra-positive-platinum-resistant
  43. Tolcher A, Hamilton E, Coleman RL. The evolving landscape of antibody-drug conjugates in gynecologic cancers. Cancer Treat Rev. 2023;116:102546. doi:10.1016/j.ctrv.2023.102546
  44. Banerjee S, Drapkin R, Richarson DL, Birrer M. Targeting NaPi2b in ovarian cancer. Cancer Treat Rev. 2023;112:102489. doi:10.1016/j.ctrv.2022.102489
Author and Disclosure Information

Ronny Drapkin, MD, PhD
Franklin Payne Associate Professor of Pathology
Department of Obstetrics and Gynecology
University of Pennsylvania Perelman School of Medicine;
Director, Ovarian Cancer Research Center
Department of Obstetrics and Gynecology
Hospital of the University of Pennsylvania
Philadelphia, PA

Ronny Drapkin, MD, PhD, has disclosed the following relevant financial relationships:
Serve(d) as a director, officer, partner, employee, advisor, consultant, or trustee for: Repare Therapeutics (scientific advisory board); VOC Health Inc (advisor).
Received income in an amount equal to or greater than $250 from: Repare Therapeutics (scientific advisory board).

Publications
Topics
Author and Disclosure Information

Ronny Drapkin, MD, PhD
Franklin Payne Associate Professor of Pathology
Department of Obstetrics and Gynecology
University of Pennsylvania Perelman School of Medicine;
Director, Ovarian Cancer Research Center
Department of Obstetrics and Gynecology
Hospital of the University of Pennsylvania
Philadelphia, PA

Ronny Drapkin, MD, PhD, has disclosed the following relevant financial relationships:
Serve(d) as a director, officer, partner, employee, advisor, consultant, or trustee for: Repare Therapeutics (scientific advisory board); VOC Health Inc (advisor).
Received income in an amount equal to or greater than $250 from: Repare Therapeutics (scientific advisory board).

Author and Disclosure Information

Ronny Drapkin, MD, PhD
Franklin Payne Associate Professor of Pathology
Department of Obstetrics and Gynecology
University of Pennsylvania Perelman School of Medicine;
Director, Ovarian Cancer Research Center
Department of Obstetrics and Gynecology
Hospital of the University of Pennsylvania
Philadelphia, PA

Ronny Drapkin, MD, PhD, has disclosed the following relevant financial relationships:
Serve(d) as a director, officer, partner, employee, advisor, consultant, or trustee for: Repare Therapeutics (scientific advisory board); VOC Health Inc (advisor).
Received income in an amount equal to or greater than $250 from: Repare Therapeutics (scientific advisory board).

Ronny Drapkin, MD, PhD
The field of ovarian cancer has experienced a paradigm shift; ovarian cancer is now known to most often arise from the fallopian tubes.1 The ovaries can act as a magnet for tumor cells that may originate elsewhere in the body. Moreover, it has been found that relatively simple risk-reducing interventions may virtually eliminate progression to invasive disease in the ovaries.1 These types of discoveries—and others—are igniting new research into novel approaches to improving outcomes for patients with ovarian cancer.

 

Incidence and Mortality

By 2040, the number of women diagnosed with ovarian cancer annually worldwide is expected to increase by 100% in low Human Development Index (HDI) countries, and by 19-28% in high HDI countries.2 The causes of this increasing incidence are likely to be multifactorial, including both hereditary and modifiable risk factors.3 In addition to increasing population size, the growing prevalence of obesity, estrogen exposures, and nulliparity are particularly pertinent as potential causes of the rising incidence of ovarian cancer in younger women. The number of ovarian cancer-related deaths is also projected to rise from about 200,000 to nearly 314,000 annually, an increase of over 50% from 2020.2,4 Although outcomes in developed regions and nations continue to improve somewhat, 5-year survival rates range from 36% to 46%.5 These outcomes are nevertheless dismal when compared with 5-year survival rates from other cancer types, such as breast cancer, which are approaching 90%.6

Principal Histotypes

The principal histotypes in ovarian cancer are epithelial in origin and include high-grade serous carcinoma, clear-cell carcinoma, endometrioid carcinoma, low-grade serous carcinoma, and mucinous carcinoma. Other rarer types are nonepithelial, ie, arising from stromal or germ cell lines.7 Incidence rates appear to be affected over time by trends such as birth rates, use of combination oral contraceptives, and menopausal hormone therapy.8 Figure 1 shows that most ovarian cancers—approximately 70%—are high-grade serous carcinoma, although in Asian countries clear cell and endometrioid carcinomas comprise a higher proportion.9

Figure 1. Major Histotypes in Ovarian Cancer
Most ovarian cancers are epithelial carcinomas. High-grade serous carcinoma is the most common, whereas the other subtypes represent 10% or fewer cases each.9

Into the Fallopian Tube

One of the most salient and dramatic discoveries of the last 2 decades has been the finding that high-grade, clear-cell, and endometrioid tumors appear to arise from tissues not normally present in the ovary.1 As a result of risk-reducing efforts to prevent serous cancers in women with genetic predisposition to develop ovarian cancer (ie, those with BRCA1 or BRCA2 mutations), it became increasingly clear that many early cancers arose in the fallopian tube,10-12 with the distal portion—the fimbria—as the most common site of origin.13-16

Figure 2 depicts the female reproductive tract, including the location of the fimbria compared with the ovaries. Moreover, lesions observed in the fallopian tube fimbria—serous tubal intraepithelial carcinomas (STICs)—were identified as precursors of ovarian cancer, with a window of 7 years between development of STIC and the beginning of an ovarian cancer.14,16

 

Figure 2. Serous Tubal Intraepithelial Carcinomas
Lesions that develop in the fallopian tube fimbria, called serous tubal intraepithelial carcinomas (STICs), have been identified as precursors of ovarian cancer.

Early Detection

Early, localized ovarian cancer is asymptomatic; by the time a patient presents with symptoms, even with nonspecific abdominal complaints, the disease is almost invariably advanced. The concept of early detection has improved both the rate of cancer diagnoses and outcomes for some malignancies, such as cervical, colorectal, breast, and lung cancers,17 but this strategy is yet to be effectively applied in ovarian cancer. A large, population-based study, for instance, yielded negative results when multimodal screening (using both measurement of CA125 blood levels and transvaginal ultrasound imaging) failed to improve survival, even though such screening was able to detect lower stage disease.18 Emerging technologies, such as liquid biopsies and uterine lavage, which seek to detect potential biomarkers (new types of blood tests) of ovarian cancer at an early stage and closer to the site of tumor origin, are being investigated and refined but are not yet ready for clinical use, particularly at the population level for screening.19

Risk-Reducing Interventions

Use of oral contraceptives has been associated with a significant reduction in risk for ovarian cancer, but the potential risks (eg, increased risk for breast cancer, increased risk for venous thromboembolism) preclude its universal recommendation.20-22 Simple removal of the fallopian tube, salpingectomy, was proposed as a potential intervention to “intercept” the progression of a STIC to cancer. Researchers recently compared simple salpingectomy with salpingo-oophorectomy as a risk-reduction procedure in carriers of BRCA 1/2 pathogenic variants after they had completed childbearing.23

These investigators proposed that later removal of the ovaries would delay menopause and would contribute to fewer/less severe symptoms, such as hot flashes, disturbed sleep, and sexual issues, as well as maintain or improve overall quality of life. The hypothesis was supported by results, which showed that patients had better menopause-related quality of life after salpingectomy than after salpingo-oophorectomy, regardless of the use of hormone replacement therapy.23 The oncologic safety of this approach was subsequently demonstrated by other studies that showed a significantly lower incidence of ovarian cancers in women who had undergone opportunistic salpingectomy.22,24,25

An international prospective trial, TUBA-WISPII, is now underway to test the hypothesis that postponement of oophorectomy after salpingectomy is non-inferior to standard salpingo-oophorectomy in terms of ovarian cancer risk for patients at high risk.26

Treatment

First-Line Therapy
Currently, there are no durable curative therapies for ovarian cancer once advanced disease has been diagnosed.

Surgery plus platinum-based chemotherapy. Most patients, even those diagnosed with advanced disease, are treated initially with debulking surgery, ideally by a gynecologic oncologist, and adjuvant chemotherapy. Most ovarian carcinomas are initially platinum-sensitive, but resistance and disease recurrence are almost inevitable. According to the National Comprehensive Cancer Network (NCCN) guidelines for ovarian cancer,27 preferred chemotherapy regimens include paclitaxel and carboplatin with or without bevacizumab, docetaxel and carboplatin, or carboplatin and liposomal doxorubicin. Numerous other regimens, combinations, and agents are included in the guidelines to help providers customize treatment plans.

Neoadjuvant vs adjuvant regimens. Neoadjuvant chemotherapy has been used for other malignancies to gauge sensitivity to systemic treatments and to improve surgical margins.28 Thus far, though, outcomes in ovarian cancer have been similar whether patients were given neoadjuvant or adjuvant treatment in the perioperative period. Individualizing these decisions based on ability to surgically resect, patient age, tumor histology, disease stage, and performance status is recommended.29

Intraperitoneal chemotherapy. Other approaches have been explored to reduce risk for micrometastases after surgery. Hyperthermic intraperitoneal chemotherapy,32 for instance, administered immediately after cytoreductive surgery was studied as a technique that might prevent some of the risks and adverse effects associated with intraperitoneal chemotherapy.31 Results showed some improvement in progression-free survival and overall survival in a subgroup of patients who underwent interval cytoreductive surgery after neoadjuvant therapy, but no differences were observed for the larger population with advanced epithelial ovarian cancer. Adverse reactions to intraperitoneal chemotherapy were also observed.

Angiogenesis inhibition. Tumors need energy and oxygen to grow. Angiogenesis is the process of new blood vessel formation that provides the tumor with nutrients. Blocking angiogenesis can thwart tumor growth and improve patient outcomes. Bevacizumab is an antiangiogenic agent that has been extensively studied for 2 decades for many cancers including ovarian carcinoma. The NCCN guidelines note that bevacizumab may be considered as part of a first-line regimen with platinum agents, as maintenance in patients with wild-type or unknown BRCA mutation status and a good response to first-line therapy, or in combination with a poly (ADP-ribose) polymerase (PARP) inhibitor in eligible patients.27

PARP inhibitors. Approximately half of all high-grade serous ovarian carcinomas exhibit some defect in the ability to repair DNA damage using the homologous recombination (HR) pathway. These tumors include those with mutations in the BRCA1, BRCA2, and other HR genes. Defects in HR make tumors more dependent on back-up DNA repair systems, including the activity of PARP. PARP inhibitors were developed to specifically target HR-deficient tumors. To date, 3 PARP inhibitors have been approved for use in ovarian cancer—olaparib, rucaparib, and niraparib. Their use has expanded from later-line use in patients with BRCA1/2-mutated tumors to include frontline maintenance regimens for women with high-grade serous and high-grade endometrioid carcinomas, as well as women with recurrent disease.32 Numerous clinical trials are ongoing to develop next-generation PARP inhibitors and to explore their efficacy in combination with chemotherapy and other targeted agents.

Resistance and Disease Progression: Second-Line and Subsequent Treatment

A number of second-line and subsequent systemic treatment regimens may be considered when primary platinum-based chemotherapy and/or maintenance are no longer effective.33,34 As emphasized by the NCCN, a clinical trial is always an appropriate option, depending on eligibility, and sometimes a second cytoreductive surgery35,36 may be considered for patients who experience radiographic and/or clinical relapse after a long disease-free interval (6+ months). Each line of treatment is associated with progressively lower response rates and shorter durations of response. According to the NCCN guidelines, as patient performance status decreases and the toxicities of each line of therapy accumulate, assessment for palliative care should be considered and discussed.27

Investigational Approaches

With the high mortality rate associated with ovarian cancer, the challenges of detecting the disease at its early stages, and the lack of therapies that can significantly extend progression-free and overall survival in patients with advanced disease, many investigators are focused on novel treatment approaches. Preclinical observations, for instance, showing synergy between ataxia telangiectasia and RAD3-related (ATR) kinase inhibitors and PARP inhibitors led researchers to initiate a phase 2 study of olaparib plus ceralasertib (an ATR inhibitor) in patients with recurrent, platinum-resistant epithelial ovarian cancer.37 No objective responses were noted, but some signals of activity were seen among patients with BRCA1 mutations.

Due to success in other malignancies, immunotherapy is also being explored. Although some promising signals were reported at 6 months when nivolumab, a PD-1 (programmed cell death protein 1) inhibitor, and ipilimumab, a cytotoxic T-lymphocyte-associated antigen 4 antibody, were combined to treat patients with platinum-resistant epithelial ovarian cancer; final results are not yet available.38

Ovarian cancer is sometimes characterized as immunologically “cold.” This description means that immune cells, especially T cells, are not able to enter the tumor and destroy the cancer cells. It also means that these tumors are not as responsive to immune-based treatments. Therefore, some researchers are examining novel alternative immunotherapy strategies, such as chimeric antigen receptor T-cell (CAR-T) therapy.39 When a CAR T-cell encounters a tumor antigen, the CAR T-cell becomes activated. Activated CAR T-cells multiply, signal to other immune cells, and ultimately kill the tumor cells. Although CAR T-cell therapy has been tremendously successful in hematologic malignancies, to date, the benefits in solid tumors have been modest.39 However, there is significant enthusiasm for novel tumor antigens that can be targeted by CAR-T therapy, including mesothelin, folate receptor, Claudin-6, B7-H3, B7-H4, HER2, CD47, and L1-CAM, among others.40

Other investigational strategies include a p53 vaccine that would enhance the patient’s immunologic response to abnormal proteins produced by a mutated p53 gene, which is the most common finding in ovarian tumors.

Although researchers are investigating many approaches to treating advanced ovarian cancer, one strategy that has been pursued in other cancer settings—development of antibody-drug conjugates (ADCs)41—has seen promising results. In the late fall of 2022, the US Food and Drug Administration granted accelerated approval for mirvetuximab soravtansine-gynx for use in patients with a specific type of type of tumor (folate receptor alpha [FRα]-positive) when platinum resistance emerges.42 A companion diagnostic assay was also approved for selecting patients with FRα-positive disease. Several other clinical trials are investigating the efficacy of targeting other ovarian tumor antigens using the ADC approach. These targets include NaPi2b, mesothelin, B7-H4, Claudin-6, and Trop-2.43,44

Progress to Come 

Progress in ovarian cancer will be made through a multipronged approach that includes interventions that may proactively “intercept” the development of cancer (eg, salpingectomy for women planning to have other simple gynecologic procedures after childbearing is complete). Although prophylactic surgeries are often undertaken by individuals at high risk for ovarian cancer because of genetic findings, such as BRCA1/2 abnormalities, even women with normal risk may consider when planning tubal ligation, removing their tubes, and other routine procedures. A substantial number of malignant tumors, and associated morbidity and mortality, may be thwarted as a result. The question of whether to treat when a STIC is detected remains to be answered.

The search for better methods of early detection continues, as local therapies for early-stage disease are invariably more effective than treatments in the advanced and/or metastatic setting.

Finally, as with certain other malignancies, even in the advanced setting, effective, often targeted, treatments can significantly prolong both progression-free and overall survival, transforming an often-lethal disease into a chronic one that allows patients to enjoy a better life expectancy with good quality of life.

Click to read more from 2023 Rare Diseases Report: Cancers

Ronny Drapkin, MD, PhD
The field of ovarian cancer has experienced a paradigm shift; ovarian cancer is now known to most often arise from the fallopian tubes.1 The ovaries can act as a magnet for tumor cells that may originate elsewhere in the body. Moreover, it has been found that relatively simple risk-reducing interventions may virtually eliminate progression to invasive disease in the ovaries.1 These types of discoveries—and others—are igniting new research into novel approaches to improving outcomes for patients with ovarian cancer.

 

Incidence and Mortality

By 2040, the number of women diagnosed with ovarian cancer annually worldwide is expected to increase by 100% in low Human Development Index (HDI) countries, and by 19-28% in high HDI countries.2 The causes of this increasing incidence are likely to be multifactorial, including both hereditary and modifiable risk factors.3 In addition to increasing population size, the growing prevalence of obesity, estrogen exposures, and nulliparity are particularly pertinent as potential causes of the rising incidence of ovarian cancer in younger women. The number of ovarian cancer-related deaths is also projected to rise from about 200,000 to nearly 314,000 annually, an increase of over 50% from 2020.2,4 Although outcomes in developed regions and nations continue to improve somewhat, 5-year survival rates range from 36% to 46%.5 These outcomes are nevertheless dismal when compared with 5-year survival rates from other cancer types, such as breast cancer, which are approaching 90%.6

Principal Histotypes

The principal histotypes in ovarian cancer are epithelial in origin and include high-grade serous carcinoma, clear-cell carcinoma, endometrioid carcinoma, low-grade serous carcinoma, and mucinous carcinoma. Other rarer types are nonepithelial, ie, arising from stromal or germ cell lines.7 Incidence rates appear to be affected over time by trends such as birth rates, use of combination oral contraceptives, and menopausal hormone therapy.8 Figure 1 shows that most ovarian cancers—approximately 70%—are high-grade serous carcinoma, although in Asian countries clear cell and endometrioid carcinomas comprise a higher proportion.9

Figure 1. Major Histotypes in Ovarian Cancer
Most ovarian cancers are epithelial carcinomas. High-grade serous carcinoma is the most common, whereas the other subtypes represent 10% or fewer cases each.9

Into the Fallopian Tube

One of the most salient and dramatic discoveries of the last 2 decades has been the finding that high-grade, clear-cell, and endometrioid tumors appear to arise from tissues not normally present in the ovary.1 As a result of risk-reducing efforts to prevent serous cancers in women with genetic predisposition to develop ovarian cancer (ie, those with BRCA1 or BRCA2 mutations), it became increasingly clear that many early cancers arose in the fallopian tube,10-12 with the distal portion—the fimbria—as the most common site of origin.13-16

Figure 2 depicts the female reproductive tract, including the location of the fimbria compared with the ovaries. Moreover, lesions observed in the fallopian tube fimbria—serous tubal intraepithelial carcinomas (STICs)—were identified as precursors of ovarian cancer, with a window of 7 years between development of STIC and the beginning of an ovarian cancer.14,16

 

Figure 2. Serous Tubal Intraepithelial Carcinomas
Lesions that develop in the fallopian tube fimbria, called serous tubal intraepithelial carcinomas (STICs), have been identified as precursors of ovarian cancer.

Early Detection

Early, localized ovarian cancer is asymptomatic; by the time a patient presents with symptoms, even with nonspecific abdominal complaints, the disease is almost invariably advanced. The concept of early detection has improved both the rate of cancer diagnoses and outcomes for some malignancies, such as cervical, colorectal, breast, and lung cancers,17 but this strategy is yet to be effectively applied in ovarian cancer. A large, population-based study, for instance, yielded negative results when multimodal screening (using both measurement of CA125 blood levels and transvaginal ultrasound imaging) failed to improve survival, even though such screening was able to detect lower stage disease.18 Emerging technologies, such as liquid biopsies and uterine lavage, which seek to detect potential biomarkers (new types of blood tests) of ovarian cancer at an early stage and closer to the site of tumor origin, are being investigated and refined but are not yet ready for clinical use, particularly at the population level for screening.19

Risk-Reducing Interventions

Use of oral contraceptives has been associated with a significant reduction in risk for ovarian cancer, but the potential risks (eg, increased risk for breast cancer, increased risk for venous thromboembolism) preclude its universal recommendation.20-22 Simple removal of the fallopian tube, salpingectomy, was proposed as a potential intervention to “intercept” the progression of a STIC to cancer. Researchers recently compared simple salpingectomy with salpingo-oophorectomy as a risk-reduction procedure in carriers of BRCA 1/2 pathogenic variants after they had completed childbearing.23

These investigators proposed that later removal of the ovaries would delay menopause and would contribute to fewer/less severe symptoms, such as hot flashes, disturbed sleep, and sexual issues, as well as maintain or improve overall quality of life. The hypothesis was supported by results, which showed that patients had better menopause-related quality of life after salpingectomy than after salpingo-oophorectomy, regardless of the use of hormone replacement therapy.23 The oncologic safety of this approach was subsequently demonstrated by other studies that showed a significantly lower incidence of ovarian cancers in women who had undergone opportunistic salpingectomy.22,24,25

An international prospective trial, TUBA-WISPII, is now underway to test the hypothesis that postponement of oophorectomy after salpingectomy is non-inferior to standard salpingo-oophorectomy in terms of ovarian cancer risk for patients at high risk.26

Treatment

First-Line Therapy
Currently, there are no durable curative therapies for ovarian cancer once advanced disease has been diagnosed.

Surgery plus platinum-based chemotherapy. Most patients, even those diagnosed with advanced disease, are treated initially with debulking surgery, ideally by a gynecologic oncologist, and adjuvant chemotherapy. Most ovarian carcinomas are initially platinum-sensitive, but resistance and disease recurrence are almost inevitable. According to the National Comprehensive Cancer Network (NCCN) guidelines for ovarian cancer,27 preferred chemotherapy regimens include paclitaxel and carboplatin with or without bevacizumab, docetaxel and carboplatin, or carboplatin and liposomal doxorubicin. Numerous other regimens, combinations, and agents are included in the guidelines to help providers customize treatment plans.

Neoadjuvant vs adjuvant regimens. Neoadjuvant chemotherapy has been used for other malignancies to gauge sensitivity to systemic treatments and to improve surgical margins.28 Thus far, though, outcomes in ovarian cancer have been similar whether patients were given neoadjuvant or adjuvant treatment in the perioperative period. Individualizing these decisions based on ability to surgically resect, patient age, tumor histology, disease stage, and performance status is recommended.29

Intraperitoneal chemotherapy. Other approaches have been explored to reduce risk for micrometastases after surgery. Hyperthermic intraperitoneal chemotherapy,32 for instance, administered immediately after cytoreductive surgery was studied as a technique that might prevent some of the risks and adverse effects associated with intraperitoneal chemotherapy.31 Results showed some improvement in progression-free survival and overall survival in a subgroup of patients who underwent interval cytoreductive surgery after neoadjuvant therapy, but no differences were observed for the larger population with advanced epithelial ovarian cancer. Adverse reactions to intraperitoneal chemotherapy were also observed.

Angiogenesis inhibition. Tumors need energy and oxygen to grow. Angiogenesis is the process of new blood vessel formation that provides the tumor with nutrients. Blocking angiogenesis can thwart tumor growth and improve patient outcomes. Bevacizumab is an antiangiogenic agent that has been extensively studied for 2 decades for many cancers including ovarian carcinoma. The NCCN guidelines note that bevacizumab may be considered as part of a first-line regimen with platinum agents, as maintenance in patients with wild-type or unknown BRCA mutation status and a good response to first-line therapy, or in combination with a poly (ADP-ribose) polymerase (PARP) inhibitor in eligible patients.27

PARP inhibitors. Approximately half of all high-grade serous ovarian carcinomas exhibit some defect in the ability to repair DNA damage using the homologous recombination (HR) pathway. These tumors include those with mutations in the BRCA1, BRCA2, and other HR genes. Defects in HR make tumors more dependent on back-up DNA repair systems, including the activity of PARP. PARP inhibitors were developed to specifically target HR-deficient tumors. To date, 3 PARP inhibitors have been approved for use in ovarian cancer—olaparib, rucaparib, and niraparib. Their use has expanded from later-line use in patients with BRCA1/2-mutated tumors to include frontline maintenance regimens for women with high-grade serous and high-grade endometrioid carcinomas, as well as women with recurrent disease.32 Numerous clinical trials are ongoing to develop next-generation PARP inhibitors and to explore their efficacy in combination with chemotherapy and other targeted agents.

Resistance and Disease Progression: Second-Line and Subsequent Treatment

A number of second-line and subsequent systemic treatment regimens may be considered when primary platinum-based chemotherapy and/or maintenance are no longer effective.33,34 As emphasized by the NCCN, a clinical trial is always an appropriate option, depending on eligibility, and sometimes a second cytoreductive surgery35,36 may be considered for patients who experience radiographic and/or clinical relapse after a long disease-free interval (6+ months). Each line of treatment is associated with progressively lower response rates and shorter durations of response. According to the NCCN guidelines, as patient performance status decreases and the toxicities of each line of therapy accumulate, assessment for palliative care should be considered and discussed.27

Investigational Approaches

With the high mortality rate associated with ovarian cancer, the challenges of detecting the disease at its early stages, and the lack of therapies that can significantly extend progression-free and overall survival in patients with advanced disease, many investigators are focused on novel treatment approaches. Preclinical observations, for instance, showing synergy between ataxia telangiectasia and RAD3-related (ATR) kinase inhibitors and PARP inhibitors led researchers to initiate a phase 2 study of olaparib plus ceralasertib (an ATR inhibitor) in patients with recurrent, platinum-resistant epithelial ovarian cancer.37 No objective responses were noted, but some signals of activity were seen among patients with BRCA1 mutations.

Due to success in other malignancies, immunotherapy is also being explored. Although some promising signals were reported at 6 months when nivolumab, a PD-1 (programmed cell death protein 1) inhibitor, and ipilimumab, a cytotoxic T-lymphocyte-associated antigen 4 antibody, were combined to treat patients with platinum-resistant epithelial ovarian cancer; final results are not yet available.38

Ovarian cancer is sometimes characterized as immunologically “cold.” This description means that immune cells, especially T cells, are not able to enter the tumor and destroy the cancer cells. It also means that these tumors are not as responsive to immune-based treatments. Therefore, some researchers are examining novel alternative immunotherapy strategies, such as chimeric antigen receptor T-cell (CAR-T) therapy.39 When a CAR T-cell encounters a tumor antigen, the CAR T-cell becomes activated. Activated CAR T-cells multiply, signal to other immune cells, and ultimately kill the tumor cells. Although CAR T-cell therapy has been tremendously successful in hematologic malignancies, to date, the benefits in solid tumors have been modest.39 However, there is significant enthusiasm for novel tumor antigens that can be targeted by CAR-T therapy, including mesothelin, folate receptor, Claudin-6, B7-H3, B7-H4, HER2, CD47, and L1-CAM, among others.40

Other investigational strategies include a p53 vaccine that would enhance the patient’s immunologic response to abnormal proteins produced by a mutated p53 gene, which is the most common finding in ovarian tumors.

Although researchers are investigating many approaches to treating advanced ovarian cancer, one strategy that has been pursued in other cancer settings—development of antibody-drug conjugates (ADCs)41—has seen promising results. In the late fall of 2022, the US Food and Drug Administration granted accelerated approval for mirvetuximab soravtansine-gynx for use in patients with a specific type of type of tumor (folate receptor alpha [FRα]-positive) when platinum resistance emerges.42 A companion diagnostic assay was also approved for selecting patients with FRα-positive disease. Several other clinical trials are investigating the efficacy of targeting other ovarian tumor antigens using the ADC approach. These targets include NaPi2b, mesothelin, B7-H4, Claudin-6, and Trop-2.43,44

Progress to Come 

Progress in ovarian cancer will be made through a multipronged approach that includes interventions that may proactively “intercept” the development of cancer (eg, salpingectomy for women planning to have other simple gynecologic procedures after childbearing is complete). Although prophylactic surgeries are often undertaken by individuals at high risk for ovarian cancer because of genetic findings, such as BRCA1/2 abnormalities, even women with normal risk may consider when planning tubal ligation, removing their tubes, and other routine procedures. A substantial number of malignant tumors, and associated morbidity and mortality, may be thwarted as a result. The question of whether to treat when a STIC is detected remains to be answered.

The search for better methods of early detection continues, as local therapies for early-stage disease are invariably more effective than treatments in the advanced and/or metastatic setting.

Finally, as with certain other malignancies, even in the advanced setting, effective, often targeted, treatments can significantly prolong both progression-free and overall survival, transforming an often-lethal disease into a chronic one that allows patients to enjoy a better life expectancy with good quality of life.

Click to read more from 2023 Rare Diseases Report: Cancers

References
  1. Karnezis AN, Cho KR, Gilks CB, Pearce CL, Huntsman DG. The disparate origins of ovarian cancers: pathogenesis and prevention strategies. Nat Rev Cancer. 2017;17(1):65-74. doi:10.1038/nrc.2016.113
  2. Cabasag CJ, Fagan PJ, Ferlay J, et al. Ovarian cancer today and tomorrow: A global assessment by world region and Human Development Index using GLOBOCAN 2020. Int J Cancer. 2022;151(9):1535-1541. doi:10.1002/ijc.34002
  3. Huang J, Chan WC, Ngai CH, et al. Worldwide burden, risk factors, and temporal trends of ovarian cancer: a global study. Cancers (Basel). 2022;14(9):2230. doi:10.3390/cancers14092230
  4. Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA Cancer J Clin. 2023;73(1):17-48. doi:10.3322/caac.21763
  5. World Ovarian Cancer Coalition. Ovarian cancer key stats. Accessed May 8, 2023. https://worldovariancancercoalition.org/about-ovarian-cancer/key-stats/
  6. American Cancer Society. Survival rates for breast cancer. Updated March 1, 2023. Accessed May 8, 2023. https://www.cancer.org/cancer/breast-cancer/understanding-a-breast-cancer-diagnosis/breast-cancer-survival-rates.html
  7. Reid BM, Permuth JB, Sellers TA. Epidemiology of ovarian cancer: a review. Cancer Biol Med. 2017;14(1):9-32. doi:10.20892/j.issn.2095-3941.2016.0084
  8. Phung MT, Pearce CL, Meza R, Jeon J. Trends of ovarian cancer incidence by histotype and race/ethnicity in the United States 1992–2019. Cancer Res Commun. 2023;3(1):1-8. doi:10.1158/2767-9764.CRC-22-0410
  9. Coburn SB, Bray F, Sherman ME, Trabert B. International patterns and trends in ovarian cancer incidence, overall and by histologic subtype. Int J Cancer. 2017;140(11):2451-2460. doi:10.1002/ijc.30676
  10. Kroeger PT Jr, Drapkin R. Pathogenesis and heterogeneity of ovarian cancer. Curr Obstet Gynecol. 2017;29(1):26-34. doi:10.1097/GCO.0000000000000340
  11. Shih lM, Wang Y, Wang TL. The origin of ovarian cancer species and precancerous landscape. Am J Pathol. 2021;191(1):26-39. doi:10.1016/j.ajpath.2020.09.006
  12. Meserve EEK, Brouwer J, Crum CP. Serous tubal intraepithelial neoplasia: the concept and its application. Mod Pathol. 2017;30(5):710-721. doi:10.1038/modpathol.2016.23
  13. Crum CP, Drapkin R, Kindelberger D, Medeiros F, Miron A, Lee Y. Lessons from BRCA: the tubal fimbria emerges as an origin for pelvic serous cancer. Clin Med Res. 2007;5(1):35-44. doi:10.3121/cmr.2007.702
  14. Wu RC, Wang P, Lin SF, et al. Genomic landscape and evolutionary trajectories of ovarian cancer precursor lesions. J Pathol. 2019;248(1):41-50. doi:10.1002/path.5219
  15. Eckert MA, Pan S, Hernandez KM, et al. Genomics of ovarian cancer progression reveals diverse metastatic trajectories including intraepithelial metastasis to the fallopian tube. Cancer Discov. 2016;6(12):1342-1351. doi:10.1158/2159-8290.CD-16-0607
  16. Labidi-Galy SI, Papp E, Hallberg D, et al. High grade serous ovarian carcinomas originate in the fallopian tube. Nat Comm. 2017;8(1):1093. doi:10.1038/s41467-017-00962-1
  17. Centers for Disease Control and Prevention; National Comprehensive Cancer Control Program (NCCP). Promoting early detection and treatment of cancer. Reviewed July 30, 2021. Accessed May 8, 2023. https://www.cdc.gov/cancer/ncccp/priorities/early-detection-treatment.htm
  18. Menon U, Gentry-Maharaj A, Burnell M, et al. Ovarian cancer population screening and mortality after long-term follow-up in the UK Collaborative Trial of Ovarian Cancer Screening (UKCTOCS): a randomised controlled trial. Lancet. 2021;397(10290):2182-2193. doi:10.1016/S0140-6736(21)00731-5
  19. Žilovic D, Ciurliene R, Sabaliauskaite R, Jarmalaitė S. Future screening prospects for ovarian cancer. Cancers (Basel). 2021;13(15):3840. doi:10.3390/cancers1315384
  20. Michels KA, Pfeiffer RM, Brinton LA, Trabert B. Modification of the associations between duration of oral contraceptive use and ovarian, endometrial, breast, and colorectal cancers. JAMA Oncol. 2018;4(4):516-521. doi:10.1001/jamaoncol.2017.4942
  21. Havrilesky LJ, Moorman PG, Lowery WJ, et al. Oral contraceptive pills as primary prevention for ovarian cancer: a systematic review and meta-analysis. Obstet Gynecol. 2013;122(1):139-147. doi:10.1097/AOG.0b013e318291c235
  22. Kotsopoulos J, Narod SA. Prophylactic salpingectomy for the prevention of ovarian cancer: who should we target? Int J Cancer. 2020;147(5):1245-1251. doi:10.1002/ijc.32916 
  23. Steenbeek MP, Harmsen MG, Hoogerbrugge N, et al. Association of salpingectomy with delayed oophorectomy versus salpingo-oophorectomy with quality of life in BRCA 1/2 pathogenic variant carriers. A nonrandomized controlled trial. JAMA Oncol. 2021;7(8):1203-1212. doi:10.1001/jamaoncol.2021.1590
  24. Hanley GE, Pearce CL, Talhouk A, et al. Outcomes from opportunistic salpingectomy for ovarian cancer prevention. JAMA Netw Open. 2022;5(2):e2147343. doi:10.1001/jamanetworkopen.2021.47343
  25. Society of Gynecologic Oncology (SGO). SGO clinical practice statement: salpingectomy for ovarian cancer prevention (SGO, November 2013). Published November 1, 2013. Accessed May 8, 2023. https://www.sgo.org/resources/sgo-clinical-practice-statement-salpingectomy-for-ovarian-cancer-prevention/
  26. Steenbeek MP, van Bommel MHD, intHout J, et al. TUBectomy with delayed oophorectomy as an alternative to risk-reducing salpingo-oophorectomy in high-risk women to assess the safety of prevention: the TUBA-WISP II study protocol [published online ahead of print, 2023 Apr 12]. Int J Gynecol Cancer. 2023;ijgc-2023-004377. doi:10.1136/ijgc-2023-004377
  27. National Comprehensive Cancer Network. NCCN clinical practice guidelines in oncology: Ovarian cancer including fallopian tube cancer and primary peritoneal cancer. Version 1.2023. December 22, 2022. Accessed May 8, 2023. https://www.nccn.org/professionals/physician_gls/pdf/ovarian.pdf
  28. Chawla A, Hunt KK, Mittendorf EA. Surgical considerations in patients receiving neoadjuvant systemic therapy. Future Oncol. 2012;8(3):239-250. doi:10.2217/fon.12.12
  29. Coleridge SL, Bryant A, Kehoe S, Morrison J. Neoadjuvant chemotherapy before surgery versus surgery followed by chemotherapy for initial treatment in advanced ovarian epithelial cancer. Cochrane Database Syst Rev. 2021;7(7):CD005343. doi:10.1002/14651858.CD005343.pub6
  30. Lim MC, Chang SJ, Park B, et al; HIPEC for Ovarian Cancer Collaborators. Survival after hyperthermic intraperitoneal chemotherapy and primary or interval cytoreductive surgery in ovarian cancer: a randomized clinical trial. JAMA Surg. 2022;157(5):374-383. doi:10.1001/jamasurg.2022.0143
  31. Walker JL, Brady MF, Wenzel L, et al. Randomized trial of intravenous versus intraperitoneal chemotherapy plus bevacizumab in advanced ovarian carcinoma: an NRG Oncology/Gynecologic Oncology Group study. J Clin Oncol. 2019;37(16):1380-1390. doi:10.1200/JCO.18.01568 
  32. Konstantinopoulos PA, Lheureux S, Moore KN. PARP inhibitors for ovarian cancer: current indications, future combinations, and novel assets in development to target DNA damage repair. Am Soc Clin Oncol Educ Book. 2020;40:1-16. doi:10.1200/EDBK_288015
  33. Markman M, Bookman MA. Second-line treatment of ovarian cancer. Oncologist. 2000;5(1):26-35. doi:10.1634/theoncologist.5-1-26
  34. Markman M. Pharmaceutical management of ovarian cancer: current status. Drugs. 2019;79(11):1231-1239. doi:10.1007/s40265-019-01158-1
  35. Bristow RE, Puri I, Chi DS. Cytoreductive surgery for recurrent ovarian cancer: a meta-analysis. Gynecol Oncol. 2009;112(1):265-274. doi:10.1016/j.ygyno.2008.08.033
  36. de Bree E, Michelakis D, Anagnostopoulou E. The current role of secondary cytoreductive surgery for recurrent ovarian cancer. Front Oncol. 2022;12:1029976. doi:10.3389/fonc.2022.1029976
  37. Shah PD, Wethington SL, Pagan C, et al. Combination ATR and PARP inhibitor (CAPRI): a phase 2 study of ceralasertib plus olaparib in patients with recurrent, platinum-resistant epithelial ovarian cancer. Gynecol Oncol. 2021;163(2): 246-253. doi:10.1016/j.ygyno.2021.08.024
  38. Borella F, Ghisoni E, Giannone G, et al. Immune checkpoint inhibitors in epithelial ovarian cancer: an overview on efficacy and future perspectives. Diagnostics (Basel). 2020;10(3):146. doi:10.3390/diagnostics10030146
  39. Wu JWY, Dand S, Doig L, et al. T-cell receptor therapy in the treatment of ovarian cancer: a mini review. Front Immunol. 2021;12:672502. doi:10.3389/fimmu.2021.672502
  40. Benard E, Casey NP, Inderberg EM, Wälchli S. SJI 2020 special issue: a catalogue of ovarian cancer targets for CAR therapy. Scand J Immunol. 2020;92(4):e12917 doi:10.1111/sji.12917
  41. Martín-Sabroso C, Lozza I, Torres-Suárez AI, Fraguas-Sánchez AI. Antibodyantineoplastic conjugates in gynecological malignancies: current status and future perspectives. Pharmaceutics. 2021;13(10):1705. doi:10.3390/pharmaceutics13101705
  42. US Food and Drug Administration. FDA grants accelerated approval to mirvetuximab soravtansine-gynx for FRα positive, platinum-resistant epithelial ovarian, fallopian tube, or peritoneal cancer. Published November 14, 2022. Accessed May 8, 2023. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-mirvetuximab-soravtansine-gynx-fra-positive-platinum-resistant
  43. Tolcher A, Hamilton E, Coleman RL. The evolving landscape of antibody-drug conjugates in gynecologic cancers. Cancer Treat Rev. 2023;116:102546. doi:10.1016/j.ctrv.2023.102546
  44. Banerjee S, Drapkin R, Richarson DL, Birrer M. Targeting NaPi2b in ovarian cancer. Cancer Treat Rev. 2023;112:102489. doi:10.1016/j.ctrv.2022.102489
References
  1. Karnezis AN, Cho KR, Gilks CB, Pearce CL, Huntsman DG. The disparate origins of ovarian cancers: pathogenesis and prevention strategies. Nat Rev Cancer. 2017;17(1):65-74. doi:10.1038/nrc.2016.113
  2. Cabasag CJ, Fagan PJ, Ferlay J, et al. Ovarian cancer today and tomorrow: A global assessment by world region and Human Development Index using GLOBOCAN 2020. Int J Cancer. 2022;151(9):1535-1541. doi:10.1002/ijc.34002
  3. Huang J, Chan WC, Ngai CH, et al. Worldwide burden, risk factors, and temporal trends of ovarian cancer: a global study. Cancers (Basel). 2022;14(9):2230. doi:10.3390/cancers14092230
  4. Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA Cancer J Clin. 2023;73(1):17-48. doi:10.3322/caac.21763
  5. World Ovarian Cancer Coalition. Ovarian cancer key stats. Accessed May 8, 2023. https://worldovariancancercoalition.org/about-ovarian-cancer/key-stats/
  6. American Cancer Society. Survival rates for breast cancer. Updated March 1, 2023. Accessed May 8, 2023. https://www.cancer.org/cancer/breast-cancer/understanding-a-breast-cancer-diagnosis/breast-cancer-survival-rates.html
  7. Reid BM, Permuth JB, Sellers TA. Epidemiology of ovarian cancer: a review. Cancer Biol Med. 2017;14(1):9-32. doi:10.20892/j.issn.2095-3941.2016.0084
  8. Phung MT, Pearce CL, Meza R, Jeon J. Trends of ovarian cancer incidence by histotype and race/ethnicity in the United States 1992–2019. Cancer Res Commun. 2023;3(1):1-8. doi:10.1158/2767-9764.CRC-22-0410
  9. Coburn SB, Bray F, Sherman ME, Trabert B. International patterns and trends in ovarian cancer incidence, overall and by histologic subtype. Int J Cancer. 2017;140(11):2451-2460. doi:10.1002/ijc.30676
  10. Kroeger PT Jr, Drapkin R. Pathogenesis and heterogeneity of ovarian cancer. Curr Obstet Gynecol. 2017;29(1):26-34. doi:10.1097/GCO.0000000000000340
  11. Shih lM, Wang Y, Wang TL. The origin of ovarian cancer species and precancerous landscape. Am J Pathol. 2021;191(1):26-39. doi:10.1016/j.ajpath.2020.09.006
  12. Meserve EEK, Brouwer J, Crum CP. Serous tubal intraepithelial neoplasia: the concept and its application. Mod Pathol. 2017;30(5):710-721. doi:10.1038/modpathol.2016.23
  13. Crum CP, Drapkin R, Kindelberger D, Medeiros F, Miron A, Lee Y. Lessons from BRCA: the tubal fimbria emerges as an origin for pelvic serous cancer. Clin Med Res. 2007;5(1):35-44. doi:10.3121/cmr.2007.702
  14. Wu RC, Wang P, Lin SF, et al. Genomic landscape and evolutionary trajectories of ovarian cancer precursor lesions. J Pathol. 2019;248(1):41-50. doi:10.1002/path.5219
  15. Eckert MA, Pan S, Hernandez KM, et al. Genomics of ovarian cancer progression reveals diverse metastatic trajectories including intraepithelial metastasis to the fallopian tube. Cancer Discov. 2016;6(12):1342-1351. doi:10.1158/2159-8290.CD-16-0607
  16. Labidi-Galy SI, Papp E, Hallberg D, et al. High grade serous ovarian carcinomas originate in the fallopian tube. Nat Comm. 2017;8(1):1093. doi:10.1038/s41467-017-00962-1
  17. Centers for Disease Control and Prevention; National Comprehensive Cancer Control Program (NCCP). Promoting early detection and treatment of cancer. Reviewed July 30, 2021. Accessed May 8, 2023. https://www.cdc.gov/cancer/ncccp/priorities/early-detection-treatment.htm
  18. Menon U, Gentry-Maharaj A, Burnell M, et al. Ovarian cancer population screening and mortality after long-term follow-up in the UK Collaborative Trial of Ovarian Cancer Screening (UKCTOCS): a randomised controlled trial. Lancet. 2021;397(10290):2182-2193. doi:10.1016/S0140-6736(21)00731-5
  19. Žilovic D, Ciurliene R, Sabaliauskaite R, Jarmalaitė S. Future screening prospects for ovarian cancer. Cancers (Basel). 2021;13(15):3840. doi:10.3390/cancers1315384
  20. Michels KA, Pfeiffer RM, Brinton LA, Trabert B. Modification of the associations between duration of oral contraceptive use and ovarian, endometrial, breast, and colorectal cancers. JAMA Oncol. 2018;4(4):516-521. doi:10.1001/jamaoncol.2017.4942
  21. Havrilesky LJ, Moorman PG, Lowery WJ, et al. Oral contraceptive pills as primary prevention for ovarian cancer: a systematic review and meta-analysis. Obstet Gynecol. 2013;122(1):139-147. doi:10.1097/AOG.0b013e318291c235
  22. Kotsopoulos J, Narod SA. Prophylactic salpingectomy for the prevention of ovarian cancer: who should we target? Int J Cancer. 2020;147(5):1245-1251. doi:10.1002/ijc.32916 
  23. Steenbeek MP, Harmsen MG, Hoogerbrugge N, et al. Association of salpingectomy with delayed oophorectomy versus salpingo-oophorectomy with quality of life in BRCA 1/2 pathogenic variant carriers. A nonrandomized controlled trial. JAMA Oncol. 2021;7(8):1203-1212. doi:10.1001/jamaoncol.2021.1590
  24. Hanley GE, Pearce CL, Talhouk A, et al. Outcomes from opportunistic salpingectomy for ovarian cancer prevention. JAMA Netw Open. 2022;5(2):e2147343. doi:10.1001/jamanetworkopen.2021.47343
  25. Society of Gynecologic Oncology (SGO). SGO clinical practice statement: salpingectomy for ovarian cancer prevention (SGO, November 2013). Published November 1, 2013. Accessed May 8, 2023. https://www.sgo.org/resources/sgo-clinical-practice-statement-salpingectomy-for-ovarian-cancer-prevention/
  26. Steenbeek MP, van Bommel MHD, intHout J, et al. TUBectomy with delayed oophorectomy as an alternative to risk-reducing salpingo-oophorectomy in high-risk women to assess the safety of prevention: the TUBA-WISP II study protocol [published online ahead of print, 2023 Apr 12]. Int J Gynecol Cancer. 2023;ijgc-2023-004377. doi:10.1136/ijgc-2023-004377
  27. National Comprehensive Cancer Network. NCCN clinical practice guidelines in oncology: Ovarian cancer including fallopian tube cancer and primary peritoneal cancer. Version 1.2023. December 22, 2022. Accessed May 8, 2023. https://www.nccn.org/professionals/physician_gls/pdf/ovarian.pdf
  28. Chawla A, Hunt KK, Mittendorf EA. Surgical considerations in patients receiving neoadjuvant systemic therapy. Future Oncol. 2012;8(3):239-250. doi:10.2217/fon.12.12
  29. Coleridge SL, Bryant A, Kehoe S, Morrison J. Neoadjuvant chemotherapy before surgery versus surgery followed by chemotherapy for initial treatment in advanced ovarian epithelial cancer. Cochrane Database Syst Rev. 2021;7(7):CD005343. doi:10.1002/14651858.CD005343.pub6
  30. Lim MC, Chang SJ, Park B, et al; HIPEC for Ovarian Cancer Collaborators. Survival after hyperthermic intraperitoneal chemotherapy and primary or interval cytoreductive surgery in ovarian cancer: a randomized clinical trial. JAMA Surg. 2022;157(5):374-383. doi:10.1001/jamasurg.2022.0143
  31. Walker JL, Brady MF, Wenzel L, et al. Randomized trial of intravenous versus intraperitoneal chemotherapy plus bevacizumab in advanced ovarian carcinoma: an NRG Oncology/Gynecologic Oncology Group study. J Clin Oncol. 2019;37(16):1380-1390. doi:10.1200/JCO.18.01568 
  32. Konstantinopoulos PA, Lheureux S, Moore KN. PARP inhibitors for ovarian cancer: current indications, future combinations, and novel assets in development to target DNA damage repair. Am Soc Clin Oncol Educ Book. 2020;40:1-16. doi:10.1200/EDBK_288015
  33. Markman M, Bookman MA. Second-line treatment of ovarian cancer. Oncologist. 2000;5(1):26-35. doi:10.1634/theoncologist.5-1-26
  34. Markman M. Pharmaceutical management of ovarian cancer: current status. Drugs. 2019;79(11):1231-1239. doi:10.1007/s40265-019-01158-1
  35. Bristow RE, Puri I, Chi DS. Cytoreductive surgery for recurrent ovarian cancer: a meta-analysis. Gynecol Oncol. 2009;112(1):265-274. doi:10.1016/j.ygyno.2008.08.033
  36. de Bree E, Michelakis D, Anagnostopoulou E. The current role of secondary cytoreductive surgery for recurrent ovarian cancer. Front Oncol. 2022;12:1029976. doi:10.3389/fonc.2022.1029976
  37. Shah PD, Wethington SL, Pagan C, et al. Combination ATR and PARP inhibitor (CAPRI): a phase 2 study of ceralasertib plus olaparib in patients with recurrent, platinum-resistant epithelial ovarian cancer. Gynecol Oncol. 2021;163(2): 246-253. doi:10.1016/j.ygyno.2021.08.024
  38. Borella F, Ghisoni E, Giannone G, et al. Immune checkpoint inhibitors in epithelial ovarian cancer: an overview on efficacy and future perspectives. Diagnostics (Basel). 2020;10(3):146. doi:10.3390/diagnostics10030146
  39. Wu JWY, Dand S, Doig L, et al. T-cell receptor therapy in the treatment of ovarian cancer: a mini review. Front Immunol. 2021;12:672502. doi:10.3389/fimmu.2021.672502
  40. Benard E, Casey NP, Inderberg EM, Wälchli S. SJI 2020 special issue: a catalogue of ovarian cancer targets for CAR therapy. Scand J Immunol. 2020;92(4):e12917 doi:10.1111/sji.12917
  41. Martín-Sabroso C, Lozza I, Torres-Suárez AI, Fraguas-Sánchez AI. Antibodyantineoplastic conjugates in gynecological malignancies: current status and future perspectives. Pharmaceutics. 2021;13(10):1705. doi:10.3390/pharmaceutics13101705
  42. US Food and Drug Administration. FDA grants accelerated approval to mirvetuximab soravtansine-gynx for FRα positive, platinum-resistant epithelial ovarian, fallopian tube, or peritoneal cancer. Published November 14, 2022. Accessed May 8, 2023. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-mirvetuximab-soravtansine-gynx-fra-positive-platinum-resistant
  43. Tolcher A, Hamilton E, Coleman RL. The evolving landscape of antibody-drug conjugates in gynecologic cancers. Cancer Treat Rev. 2023;116:102546. doi:10.1016/j.ctrv.2023.102546
  44. Banerjee S, Drapkin R, Richarson DL, Birrer M. Targeting NaPi2b in ovarian cancer. Cancer Treat Rev. 2023;112:102489. doi:10.1016/j.ctrv.2022.102489
Publications
Publications
Topics
Article Type
Display Headline
Progress in Ovarian Cancer: Discovery of Fallopian Tube Involvement
Display Headline
Progress in Ovarian Cancer: Discovery of Fallopian Tube Involvement
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Fri, 06/02/2023 - 11:45
Un-Gate On Date
Fri, 06/02/2023 - 11:45
Use ProPublica
CFC Schedule Remove Status
Fri, 06/02/2023 - 11:45
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Time for mental stress testing in ANOCA?

Article Type
Changed
Tue, 06/27/2023 - 12:04

Among women with angina without obstructive coronary artery disease (ANOCA), mental stress induced a greater degree of myocardial ischemia than among those without ANOCA, new results show.

Further analysis in the small study suggested that mental stress–induced myocardial ischemia (MSIMI) was not statistically related to coronary microvascular dysfunction (CMD).

“Since the findings do not support a correlation between MSIMI and CMD, which has been a widely accepted mechanistic explanation of ANOCA, routine mental stress testing in patients with ANOCA seems necessary,” researchers led by Qingshan Geng, MD, PhD, of Shenzhen People’s Hospital, Guangdong, China, conclude in a report published online in the Journal of the American College of Cardiology.

Dr. Geng said in an interview that the use of virtual reality devices to administer mental stress tests “ensures standardized experimental procedures, with each participant receiving an objectively equivalent level of stress load.

“The immersive experience provided by VR lowers the environmental requirements for the test,” he noted. “Furthermore, the application of VR reduces the workload of personnel responsible for inducing mental stress, simplifying the experimental process.”

The team also developed a mobile app that enables remote monitoring of participants’ visual experiences during PET/CT scans and facilitates communication, he added.
 

Mental stress testing and meds?

Both ANOCA and MSIMI in patients with coronary artery disease disproportionately affect women and are associated with poor cardiovascular prognosis, the researchers write.

“However, the role of MSIMI and the exact influence of mental stress in ANOCA have not previously been studied,” they point out.

For this investigation, 84 women with ANOCA and 42 age-matched controls underwent three mental stress challenges delivered via VR.

Tests included mental arithmetic, making a public speech describing a recent emotionally upsetting event, and a task-modified Stroop test, in which participants were asked to say the color in which the word appears, not the color that the word names. For example, if the word “yellow” appears in blue type, blue would be the correct answer.

An adenosine stress test was given 5-8 minutes after the mental stress challenges started, and cardiac PET/CT was used to examine myocardial blood flow and perfusion.

The investigators report that women with ANOCA had a much higher rate of MSIMI (42.9%), compared with control participants (one patient; 2.4%). They also had a higher proportion of coronary microvascular dysfunction (CMD; 24.6% vs. 8.6%), but the occurrence of MSIMI and CMD was not related, the authors note.

Consistent with previous studies, “we observed that CMD is more prevalent in ANOCA women than the age-matched healthy individuals. MSIMI rate, however, was notably higher than the rate of CMD in our female ANOCA population,” they write. “The lack of a significant association between MSIMI and CMD indicates the mechanisms of MSIMI cannot be well explained by the adenosine-induced CMD.”

Dr. Geng suggested that ANOCA patients may benefit from treatment with escitalopram.

“Compelling evidence” from the REMIT randomized, placebo-controlled trial validates the efficacy of the drug as an MSIMI treatment, he said.
 

Sample size too small?

Asked for comment on the findings, Viola Vaccarino, MD, PhD, Wilton Looney Distinguished Professor of Cardiovascular Research at Emory University’s Rollins School of Public Health and a professor in the university’s School of Medicine, Atlanta, said she disagreed with several aspects of this study and the investigators’ conclusions.

Although the study suggests that MSIMI is prevalent among women with ANOCA, “the sample size was too small to make any definite conclusions,” she said in an interview.

“In fact,” she said, “I do not agree with the authors’ conclusions that MSIMI and CMD were not related, based on the data presented, even though the P value was not significant.”

In addition, more research is needed before screening can be recommended, she said. “The effectiveness of this testing modality in this population should be demonstrated first.”

Furthermore, she added, “an established treatment for MSIMI has yet to be tested in large, controlled trials, which limits the potential clinical benefit that may result from this [screening] test.”

For now, to ameliorate potential MSIMI in women with ANOCA, Dr. Vaccarino recommends behavioral modalities or stress-reduction management techniques, including biofeedback, meditation, breathing exercises, and “just plain regular physical activity,” rather than the use of psychotropic medications.

Dr. Vaccarino’s team has a study underway that builds on earlier work involving more than 900 participants, which showed that MSIMI was significantly associated with an increased risk of cardiovascular death or nonfatal myocardial infarction (hazard ratio, 2.5).

The ongoing study, which investigates the link between emotional stress and heart disease in men and women, should be completed in about 3 years, she said.
 

Microvascular disease or spasm?

Leslie Cho, MD, chair of the American College of Cardiology’s Cardiovascular Disease in Women Committee, director of the Cleveland Clinic’s Women’s Cardiovascular Center, and professor of medicine at Cleveland Clinic Lerner School of Medicine and Case Western Reserve Medical School, commented on the mental stress–heart connection and mental stress testing for this article.

A “very big flaw” of the JACC study, she said, is that although PET testing can detect microvascular disease, it cannot detect microvascular spasm.

PET can show the coronary flow reserve, “which is a nice way to assess microvascular dysfunction,” she acknowledged, “but it really can’t tell microvascular spasm, because adenosine works in a different pathway than acetylcholine – and I think it’s important for people to have the right diagnosis.

“We do physiologic testing to distinguish the two conditions,” she noted. “We do the gold standard, which is the cath lab.”

“The problem with women with chest pain for years is that they get a stress test, they get a cath, and everything’s normal. Then they get blown off as anxious or whatever.”

Clinicians should conduct the gold standard workup – provocative physiologic testing – for these women who continue to have chest pain when results of other tests are negative, she said. “The test used to be very cumbersome, but today, we have systems that make it super easy to use and to distinguish microvascular disease and microvascular spasm.”

Importantly, she added, physiologic testing should be performed when women are off therapy – something that doesn’t always happen in the clinic.

Regarding treatment, she added, “if you’re having emotional stress, the answer is not another medicine. The answer is cognitive-behavioral therapy or another behavioral intervention to overcome anxiety.”
 

Tune in and advocate

What can clinicians do for women with ANOCA after testing reveals no significant coronary artery disease or microvascular spasms?

“Very often, it’s a matter of the doctor listening and responding to the patient,” Johanna Contreras, MD, a cardiologist at Mount Sinai Hospital, New York, said in an intereview.

In her practice, Dr. Contreras sees highly stressed women on a daily basis. Many of her patients are women from diverse racial/ethnic groups, often of lower socioeconomic status, who are heads of households, work more than one job, and experience other major stressors.

“My message to clinicians is: don’t give up on a woman just because you looked at the arteries and couldn’t find anything specific. If she keeps coming back with the same symptom, it’s important to address it,” she said. “Maybe it isn’t the symptom. Maybe she needs to talk about her situation, about the physiological and psychosocial factors contributing to the symptom that a test alone won’t reveal.”

Regarding cardiovascular spasms that are identified through physiologic testing, she said, “I don’t know that medications such as SSRIs [selective serotonin reuptake inhibitors] are going to change anything. But many things can be changed by listening or helping the patient to stop and think about her mental health.”

Following up with a referral to a therapist can help, she said; “Take away the mental health stigma by telling the patient that the referral is simply to help her cope.”

Dr. Contreras urges clinicians to be advocates for such patients. If an insurance company says it will cover only three therapy sessions, “tell them that three appointments are not enough” to address multiple issues.

“If we invest money in helping patients identify and cope with these issues, we are likely to get better long-term outcomes, rather than having that woman come into the emergency department with chest pain over and over and doing 20,000 tests that are going to show exactly the same thing,” Dr. Contreras concluded.

Dr. Geng’s study was supported by the High-Level Hospital Construction Project of Guangdong Provincial People’s Hospital, by a grant from Guangdong Provincial Bureau of Traditional Chinese Medicine, and by a grant from Guangdong Medical Science and Technology Research Foundation. The authors, Dr. Vaccarino, Dr. Contreras, and Dr. Cho report no relevant financial relationships.

A version of this article originally appeared on Medscape.com.

Publications
Topics
Sections

Among women with angina without obstructive coronary artery disease (ANOCA), mental stress induced a greater degree of myocardial ischemia than among those without ANOCA, new results show.

Further analysis in the small study suggested that mental stress–induced myocardial ischemia (MSIMI) was not statistically related to coronary microvascular dysfunction (CMD).

“Since the findings do not support a correlation between MSIMI and CMD, which has been a widely accepted mechanistic explanation of ANOCA, routine mental stress testing in patients with ANOCA seems necessary,” researchers led by Qingshan Geng, MD, PhD, of Shenzhen People’s Hospital, Guangdong, China, conclude in a report published online in the Journal of the American College of Cardiology.

Dr. Geng said in an interview that the use of virtual reality devices to administer mental stress tests “ensures standardized experimental procedures, with each participant receiving an objectively equivalent level of stress load.

“The immersive experience provided by VR lowers the environmental requirements for the test,” he noted. “Furthermore, the application of VR reduces the workload of personnel responsible for inducing mental stress, simplifying the experimental process.”

The team also developed a mobile app that enables remote monitoring of participants’ visual experiences during PET/CT scans and facilitates communication, he added.
 

Mental stress testing and meds?

Both ANOCA and MSIMI in patients with coronary artery disease disproportionately affect women and are associated with poor cardiovascular prognosis, the researchers write.

“However, the role of MSIMI and the exact influence of mental stress in ANOCA have not previously been studied,” they point out.

For this investigation, 84 women with ANOCA and 42 age-matched controls underwent three mental stress challenges delivered via VR.

Tests included mental arithmetic, making a public speech describing a recent emotionally upsetting event, and a task-modified Stroop test, in which participants were asked to say the color in which the word appears, not the color that the word names. For example, if the word “yellow” appears in blue type, blue would be the correct answer.

An adenosine stress test was given 5-8 minutes after the mental stress challenges started, and cardiac PET/CT was used to examine myocardial blood flow and perfusion.

The investigators report that women with ANOCA had a much higher rate of MSIMI (42.9%), compared with control participants (one patient; 2.4%). They also had a higher proportion of coronary microvascular dysfunction (CMD; 24.6% vs. 8.6%), but the occurrence of MSIMI and CMD was not related, the authors note.

Consistent with previous studies, “we observed that CMD is more prevalent in ANOCA women than the age-matched healthy individuals. MSIMI rate, however, was notably higher than the rate of CMD in our female ANOCA population,” they write. “The lack of a significant association between MSIMI and CMD indicates the mechanisms of MSIMI cannot be well explained by the adenosine-induced CMD.”

Dr. Geng suggested that ANOCA patients may benefit from treatment with escitalopram.

“Compelling evidence” from the REMIT randomized, placebo-controlled trial validates the efficacy of the drug as an MSIMI treatment, he said.
 

Sample size too small?

Asked for comment on the findings, Viola Vaccarino, MD, PhD, Wilton Looney Distinguished Professor of Cardiovascular Research at Emory University’s Rollins School of Public Health and a professor in the university’s School of Medicine, Atlanta, said she disagreed with several aspects of this study and the investigators’ conclusions.

Although the study suggests that MSIMI is prevalent among women with ANOCA, “the sample size was too small to make any definite conclusions,” she said in an interview.

“In fact,” she said, “I do not agree with the authors’ conclusions that MSIMI and CMD were not related, based on the data presented, even though the P value was not significant.”

In addition, more research is needed before screening can be recommended, she said. “The effectiveness of this testing modality in this population should be demonstrated first.”

Furthermore, she added, “an established treatment for MSIMI has yet to be tested in large, controlled trials, which limits the potential clinical benefit that may result from this [screening] test.”

For now, to ameliorate potential MSIMI in women with ANOCA, Dr. Vaccarino recommends behavioral modalities or stress-reduction management techniques, including biofeedback, meditation, breathing exercises, and “just plain regular physical activity,” rather than the use of psychotropic medications.

Dr. Vaccarino’s team has a study underway that builds on earlier work involving more than 900 participants, which showed that MSIMI was significantly associated with an increased risk of cardiovascular death or nonfatal myocardial infarction (hazard ratio, 2.5).

The ongoing study, which investigates the link between emotional stress and heart disease in men and women, should be completed in about 3 years, she said.
 

Microvascular disease or spasm?

Leslie Cho, MD, chair of the American College of Cardiology’s Cardiovascular Disease in Women Committee, director of the Cleveland Clinic’s Women’s Cardiovascular Center, and professor of medicine at Cleveland Clinic Lerner School of Medicine and Case Western Reserve Medical School, commented on the mental stress–heart connection and mental stress testing for this article.

A “very big flaw” of the JACC study, she said, is that although PET testing can detect microvascular disease, it cannot detect microvascular spasm.

PET can show the coronary flow reserve, “which is a nice way to assess microvascular dysfunction,” she acknowledged, “but it really can’t tell microvascular spasm, because adenosine works in a different pathway than acetylcholine – and I think it’s important for people to have the right diagnosis.

“We do physiologic testing to distinguish the two conditions,” she noted. “We do the gold standard, which is the cath lab.”

“The problem with women with chest pain for years is that they get a stress test, they get a cath, and everything’s normal. Then they get blown off as anxious or whatever.”

Clinicians should conduct the gold standard workup – provocative physiologic testing – for these women who continue to have chest pain when results of other tests are negative, she said. “The test used to be very cumbersome, but today, we have systems that make it super easy to use and to distinguish microvascular disease and microvascular spasm.”

Importantly, she added, physiologic testing should be performed when women are off therapy – something that doesn’t always happen in the clinic.

Regarding treatment, she added, “if you’re having emotional stress, the answer is not another medicine. The answer is cognitive-behavioral therapy or another behavioral intervention to overcome anxiety.”
 

Tune in and advocate

What can clinicians do for women with ANOCA after testing reveals no significant coronary artery disease or microvascular spasms?

“Very often, it’s a matter of the doctor listening and responding to the patient,” Johanna Contreras, MD, a cardiologist at Mount Sinai Hospital, New York, said in an intereview.

In her practice, Dr. Contreras sees highly stressed women on a daily basis. Many of her patients are women from diverse racial/ethnic groups, often of lower socioeconomic status, who are heads of households, work more than one job, and experience other major stressors.

“My message to clinicians is: don’t give up on a woman just because you looked at the arteries and couldn’t find anything specific. If she keeps coming back with the same symptom, it’s important to address it,” she said. “Maybe it isn’t the symptom. Maybe she needs to talk about her situation, about the physiological and psychosocial factors contributing to the symptom that a test alone won’t reveal.”

Regarding cardiovascular spasms that are identified through physiologic testing, she said, “I don’t know that medications such as SSRIs [selective serotonin reuptake inhibitors] are going to change anything. But many things can be changed by listening or helping the patient to stop and think about her mental health.”

Following up with a referral to a therapist can help, she said; “Take away the mental health stigma by telling the patient that the referral is simply to help her cope.”

Dr. Contreras urges clinicians to be advocates for such patients. If an insurance company says it will cover only three therapy sessions, “tell them that three appointments are not enough” to address multiple issues.

“If we invest money in helping patients identify and cope with these issues, we are likely to get better long-term outcomes, rather than having that woman come into the emergency department with chest pain over and over and doing 20,000 tests that are going to show exactly the same thing,” Dr. Contreras concluded.

Dr. Geng’s study was supported by the High-Level Hospital Construction Project of Guangdong Provincial People’s Hospital, by a grant from Guangdong Provincial Bureau of Traditional Chinese Medicine, and by a grant from Guangdong Medical Science and Technology Research Foundation. The authors, Dr. Vaccarino, Dr. Contreras, and Dr. Cho report no relevant financial relationships.

A version of this article originally appeared on Medscape.com.

Among women with angina without obstructive coronary artery disease (ANOCA), mental stress induced a greater degree of myocardial ischemia than among those without ANOCA, new results show.

Further analysis in the small study suggested that mental stress–induced myocardial ischemia (MSIMI) was not statistically related to coronary microvascular dysfunction (CMD).

“Since the findings do not support a correlation between MSIMI and CMD, which has been a widely accepted mechanistic explanation of ANOCA, routine mental stress testing in patients with ANOCA seems necessary,” researchers led by Qingshan Geng, MD, PhD, of Shenzhen People’s Hospital, Guangdong, China, conclude in a report published online in the Journal of the American College of Cardiology.

Dr. Geng said in an interview that the use of virtual reality devices to administer mental stress tests “ensures standardized experimental procedures, with each participant receiving an objectively equivalent level of stress load.

“The immersive experience provided by VR lowers the environmental requirements for the test,” he noted. “Furthermore, the application of VR reduces the workload of personnel responsible for inducing mental stress, simplifying the experimental process.”

The team also developed a mobile app that enables remote monitoring of participants’ visual experiences during PET/CT scans and facilitates communication, he added.
 

Mental stress testing and meds?

Both ANOCA and MSIMI in patients with coronary artery disease disproportionately affect women and are associated with poor cardiovascular prognosis, the researchers write.

“However, the role of MSIMI and the exact influence of mental stress in ANOCA have not previously been studied,” they point out.

For this investigation, 84 women with ANOCA and 42 age-matched controls underwent three mental stress challenges delivered via VR.

Tests included mental arithmetic, making a public speech describing a recent emotionally upsetting event, and a task-modified Stroop test, in which participants were asked to say the color in which the word appears, not the color that the word names. For example, if the word “yellow” appears in blue type, blue would be the correct answer.

An adenosine stress test was given 5-8 minutes after the mental stress challenges started, and cardiac PET/CT was used to examine myocardial blood flow and perfusion.

The investigators report that women with ANOCA had a much higher rate of MSIMI (42.9%), compared with control participants (one patient; 2.4%). They also had a higher proportion of coronary microvascular dysfunction (CMD; 24.6% vs. 8.6%), but the occurrence of MSIMI and CMD was not related, the authors note.

Consistent with previous studies, “we observed that CMD is more prevalent in ANOCA women than the age-matched healthy individuals. MSIMI rate, however, was notably higher than the rate of CMD in our female ANOCA population,” they write. “The lack of a significant association between MSIMI and CMD indicates the mechanisms of MSIMI cannot be well explained by the adenosine-induced CMD.”

Dr. Geng suggested that ANOCA patients may benefit from treatment with escitalopram.

“Compelling evidence” from the REMIT randomized, placebo-controlled trial validates the efficacy of the drug as an MSIMI treatment, he said.
 

Sample size too small?

Asked for comment on the findings, Viola Vaccarino, MD, PhD, Wilton Looney Distinguished Professor of Cardiovascular Research at Emory University’s Rollins School of Public Health and a professor in the university’s School of Medicine, Atlanta, said she disagreed with several aspects of this study and the investigators’ conclusions.

Although the study suggests that MSIMI is prevalent among women with ANOCA, “the sample size was too small to make any definite conclusions,” she said in an interview.

“In fact,” she said, “I do not agree with the authors’ conclusions that MSIMI and CMD were not related, based on the data presented, even though the P value was not significant.”

In addition, more research is needed before screening can be recommended, she said. “The effectiveness of this testing modality in this population should be demonstrated first.”

Furthermore, she added, “an established treatment for MSIMI has yet to be tested in large, controlled trials, which limits the potential clinical benefit that may result from this [screening] test.”

For now, to ameliorate potential MSIMI in women with ANOCA, Dr. Vaccarino recommends behavioral modalities or stress-reduction management techniques, including biofeedback, meditation, breathing exercises, and “just plain regular physical activity,” rather than the use of psychotropic medications.

Dr. Vaccarino’s team has a study underway that builds on earlier work involving more than 900 participants, which showed that MSIMI was significantly associated with an increased risk of cardiovascular death or nonfatal myocardial infarction (hazard ratio, 2.5).

The ongoing study, which investigates the link between emotional stress and heart disease in men and women, should be completed in about 3 years, she said.
 

Microvascular disease or spasm?

Leslie Cho, MD, chair of the American College of Cardiology’s Cardiovascular Disease in Women Committee, director of the Cleveland Clinic’s Women’s Cardiovascular Center, and professor of medicine at Cleveland Clinic Lerner School of Medicine and Case Western Reserve Medical School, commented on the mental stress–heart connection and mental stress testing for this article.

A “very big flaw” of the JACC study, she said, is that although PET testing can detect microvascular disease, it cannot detect microvascular spasm.

PET can show the coronary flow reserve, “which is a nice way to assess microvascular dysfunction,” she acknowledged, “but it really can’t tell microvascular spasm, because adenosine works in a different pathway than acetylcholine – and I think it’s important for people to have the right diagnosis.

“We do physiologic testing to distinguish the two conditions,” she noted. “We do the gold standard, which is the cath lab.”

“The problem with women with chest pain for years is that they get a stress test, they get a cath, and everything’s normal. Then they get blown off as anxious or whatever.”

Clinicians should conduct the gold standard workup – provocative physiologic testing – for these women who continue to have chest pain when results of other tests are negative, she said. “The test used to be very cumbersome, but today, we have systems that make it super easy to use and to distinguish microvascular disease and microvascular spasm.”

Importantly, she added, physiologic testing should be performed when women are off therapy – something that doesn’t always happen in the clinic.

Regarding treatment, she added, “if you’re having emotional stress, the answer is not another medicine. The answer is cognitive-behavioral therapy or another behavioral intervention to overcome anxiety.”
 

Tune in and advocate

What can clinicians do for women with ANOCA after testing reveals no significant coronary artery disease or microvascular spasms?

“Very often, it’s a matter of the doctor listening and responding to the patient,” Johanna Contreras, MD, a cardiologist at Mount Sinai Hospital, New York, said in an intereview.

In her practice, Dr. Contreras sees highly stressed women on a daily basis. Many of her patients are women from diverse racial/ethnic groups, often of lower socioeconomic status, who are heads of households, work more than one job, and experience other major stressors.

“My message to clinicians is: don’t give up on a woman just because you looked at the arteries and couldn’t find anything specific. If she keeps coming back with the same symptom, it’s important to address it,” she said. “Maybe it isn’t the symptom. Maybe she needs to talk about her situation, about the physiological and psychosocial factors contributing to the symptom that a test alone won’t reveal.”

Regarding cardiovascular spasms that are identified through physiologic testing, she said, “I don’t know that medications such as SSRIs [selective serotonin reuptake inhibitors] are going to change anything. But many things can be changed by listening or helping the patient to stop and think about her mental health.”

Following up with a referral to a therapist can help, she said; “Take away the mental health stigma by telling the patient that the referral is simply to help her cope.”

Dr. Contreras urges clinicians to be advocates for such patients. If an insurance company says it will cover only three therapy sessions, “tell them that three appointments are not enough” to address multiple issues.

“If we invest money in helping patients identify and cope with these issues, we are likely to get better long-term outcomes, rather than having that woman come into the emergency department with chest pain over and over and doing 20,000 tests that are going to show exactly the same thing,” Dr. Contreras concluded.

Dr. Geng’s study was supported by the High-Level Hospital Construction Project of Guangdong Provincial People’s Hospital, by a grant from Guangdong Provincial Bureau of Traditional Chinese Medicine, and by a grant from Guangdong Medical Science and Technology Research Foundation. The authors, Dr. Vaccarino, Dr. Contreras, and Dr. Cho report no relevant financial relationships.

A version of this article originally appeared on Medscape.com.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

SAFE algorithm detects liver disease in general population

Article Type
Changed
Tue, 06/27/2023 - 08:29

VIENNA – An algorithm, the Steatosis-Associated Fibrosis Estimator (SAFE), was developed to detect clinically significant fibrosis in patients with nonalcoholic fatty liver disease (NAFLD). It is effective at detecting chronic liver disease from all causes with or without NAFLD in the general population, according the results of a U.S. population-based study. The algorithm was designed for use in primary care to help slow the steep rise in liver disease burden.

On the basis of the SAFE score, 61.3% of participants were at low risk for clinically significant fibrosis; 11.2% were at high risk; and 27.5% were at intermediate risk. Upon validation, very few of the low-risk participants had liver fibrosis, while nearly a third of those with a high-risk score had clinically significant fibrosis. In addition, a high percentage of the patients with high-risk SAFE scores had viral hepatitis and elevations in ferritin level.

“This is the first time that there has been a test that provides a score to detect low-risk liver disease in primary care,” said Ray Kim, MD, from Stanford (Calif.) University, senior investigator, who was speaking to this news organization at the annual International Liver Congress sponsored by the European Association for the Study of the Liver

“Primary care doctors currently detect liver disease through a serendipitous abnormal finding on ultrasound or blood tests that detect elevated transaminases, and then the patient is referred to a hepatologist, who figures out what is really going on,” said Dr. Kim.

“This approach is limited, so we need to get SAFE into primary care so these doctors can automatically calculate their scores, and if the patient is over 100 [high risk of chronic liver disease], then they need help [referral to a hepatologist].”
 

Liver deaths sharply rising

Public health data show that more people are dying of liver disease today than previously. Deaths in the United States have doubled over the past 20 years, said Dr. Kim. “If our mission is to help these patients and prevent death, [things are] moving in the wrong direction.”

He stressed that in order to change the direction, “primary care doctors need to engage with the issue and have appropriate tools to identify people with liver disease.”

Most often, the reason for this rise in deaths is that cases are being diagnosed at advanced stages of disease in which reversibility is limited, he added. “We want to move upstream where people might have early-stage disease and where we can intervene and make a difference.”

In an effort to help earlier detection of liver disease, the SAFE score was developed and validated by Dr. Kim and his colleagues to detect clinically significant (greater than stage 2) fibrosis in patients with NAFLD in primary care. The score is based upon age, body mass index, diabetes, platelet level, aspartate and alanine aminotransferase levels, and globulin level. A score of less than zero signifies that a patient is at low risk for liver fibrosis, while a score greater than 100 signifies a high risk of fibrosis. A score between 0 and 99 denotes intermediate risk of fibrosis.

“Unlike other noninvasive tests that detect advanced fibrosis, this one detects early-stage fibrosis. We’ve shown that the SAFE estimator is better than all other blood-based markers,” explained Dr. Kim.
 

 

 

Applying SAFE to the general population

In the study presented here at EASL, Dr. Kim aimed to expand the horizon for SAFE testing to the U.S. general population and to assess whether SAFE was effective in screening for chronic liver disease regardless of steatosis of the liver.

Together with first author Nakia Chung, MD, also from Stanford University, Dr. Kim applied the SAFE score to data from 7,156 participants of the National Health and Nutrition Examination Survey (NHANES) for 2017-2020. NHANES is representative of the noninstitutionalized, civilian population of the United States. It includes broad demographic, clinical, and laboratory data, including transient elastography data. FibroScans were first used in 2017, so the investigators had 3 years of FibroScan data with which to validate the score.

The researchers extrapolated the NHANES sample data to the U.S. population. They found that the proportion of adults with steatosis (CAP score > 274 dB/m) and significant fibrosis (LSM > 8.0 kPa) was 42.7% (95% confidence interval, 41.0%- 44.3%) and 8.9% (7.6%-10.2%), respectively. In addition, 11.3% (10.2%-12.5%) of the adult U.S. population demonstrated a significant amount of alcohol use, 2.3% (1.4%-3.3%) showed evidence of hepatitis B or C, and 5.4% (4.6%-6.2%) had elevated serum ferritin levels.

The researchers then stratified the patients according to previously defined SAFE tiers of low, intermediate, and high risk and projected findings to the U.S. general population.

“When we applied our score to the general population, we found multiple abnormalities in the high-risk groups [SAFE >100] having Fibroscan data that are consistent with stage 2 or higher fibrosis regardless of etiology, “Dr. Kim pointed out.

Results also showed that very few patients with SAFE less than 0 had liver fibrosis (4% among those with liver stiffness measure [LSM] > 8kPa, and 0.8% with LSM > 12kPa). Among those with SAFE > 100, nearly a third (31.5%) had LSM of > 8kPa, and 16.5% had LSM > 12kPa.

In addition to fibrosis, liver abnormalities were common among patients with SAFE greater than 100, including steatosis (68.0%), viral hepatitis (7.0%), and abnormal ferritin levels (12.9%); 10.8% of these patients used alcohol.

“Right now, some patients are referred, but on examination and FibroScan, they might actually be okay, so it it’s a waste of time and money for everyone. We can preempt all of this by doing a blood test and focusing on those people who really need a scan,” said Dr. Kim.

The researcher is now working with primary care colleagues to help further develop and integrate SAFE into the primary care setting.
 

Fibrosis score in patients with metabolic dysfunction

Also presenting at the same session on population health was Willem Pieter Brouwer, MD, PhD, from Erasmus University Medical Center, Rotterdam, the Netherlands. He reported results of a validation study of a new risk score – the Metabolic Dysfunction-Associated Fibrosis–5 (MAF-5) score – for use for people with metabolic dysfunction who are recommended for screening for liver fibrosis.

“We believe the MAF-5 score may be a good alternative to the FIB-4 [a liver fibrosis biomarker] for use in the referral pathway for liver health evaluation,” remarked Dr. Brouwer. “The clinical practice guidelines recommend using FIB-4 scores, but these have a poor-moderate performance in the population setting.

“We developed and validated our score in a population of 5,500 from the NHANES 2017-2020 cycle and validated the score in populations from Rotterdam, which is a cohort of elderly participants, and in fibrosis among patients with biopsy-proven NAFLD from Colombia and Belgium,” he explained.

He also validated the score against different existing scoring systems and different methods of measuring liver stiffness and validated it for prognostic use to predict all-cause mortality in the NHANES III cohort.

Dr. Brouwer removed age as a factor of his new MAF-5 score; the score is thus stable for patients of all ages and is suitable for detecting liver disease in younger patients. “This is very important because these patients are currently underserved and have the most years of life to win.”

Referring to the SAFE score discussed by Dr. Kim, as well as other scores, he said, “The FIB-4, SAFE, and NFS [NAFLD fibrosis score] all include age in the scores, which causes problems and limitations in aging populations, as more and more patients will be referred due to an increasing score. Hence, the elderly are mostly all referred for liver checkups.”

Dr. Kim and Dr. Brouwer have disclosed no relevant financial relationships.

A version of this article first appeared on Medscape.com.

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

VIENNA – An algorithm, the Steatosis-Associated Fibrosis Estimator (SAFE), was developed to detect clinically significant fibrosis in patients with nonalcoholic fatty liver disease (NAFLD). It is effective at detecting chronic liver disease from all causes with or without NAFLD in the general population, according the results of a U.S. population-based study. The algorithm was designed for use in primary care to help slow the steep rise in liver disease burden.

On the basis of the SAFE score, 61.3% of participants were at low risk for clinically significant fibrosis; 11.2% were at high risk; and 27.5% were at intermediate risk. Upon validation, very few of the low-risk participants had liver fibrosis, while nearly a third of those with a high-risk score had clinically significant fibrosis. In addition, a high percentage of the patients with high-risk SAFE scores had viral hepatitis and elevations in ferritin level.

“This is the first time that there has been a test that provides a score to detect low-risk liver disease in primary care,” said Ray Kim, MD, from Stanford (Calif.) University, senior investigator, who was speaking to this news organization at the annual International Liver Congress sponsored by the European Association for the Study of the Liver

“Primary care doctors currently detect liver disease through a serendipitous abnormal finding on ultrasound or blood tests that detect elevated transaminases, and then the patient is referred to a hepatologist, who figures out what is really going on,” said Dr. Kim.

“This approach is limited, so we need to get SAFE into primary care so these doctors can automatically calculate their scores, and if the patient is over 100 [high risk of chronic liver disease], then they need help [referral to a hepatologist].”
 

Liver deaths sharply rising

Public health data show that more people are dying of liver disease today than previously. Deaths in the United States have doubled over the past 20 years, said Dr. Kim. “If our mission is to help these patients and prevent death, [things are] moving in the wrong direction.”

He stressed that in order to change the direction, “primary care doctors need to engage with the issue and have appropriate tools to identify people with liver disease.”

Most often, the reason for this rise in deaths is that cases are being diagnosed at advanced stages of disease in which reversibility is limited, he added. “We want to move upstream where people might have early-stage disease and where we can intervene and make a difference.”

In an effort to help earlier detection of liver disease, the SAFE score was developed and validated by Dr. Kim and his colleagues to detect clinically significant (greater than stage 2) fibrosis in patients with NAFLD in primary care. The score is based upon age, body mass index, diabetes, platelet level, aspartate and alanine aminotransferase levels, and globulin level. A score of less than zero signifies that a patient is at low risk for liver fibrosis, while a score greater than 100 signifies a high risk of fibrosis. A score between 0 and 99 denotes intermediate risk of fibrosis.

“Unlike other noninvasive tests that detect advanced fibrosis, this one detects early-stage fibrosis. We’ve shown that the SAFE estimator is better than all other blood-based markers,” explained Dr. Kim.
 

 

 

Applying SAFE to the general population

In the study presented here at EASL, Dr. Kim aimed to expand the horizon for SAFE testing to the U.S. general population and to assess whether SAFE was effective in screening for chronic liver disease regardless of steatosis of the liver.

Together with first author Nakia Chung, MD, also from Stanford University, Dr. Kim applied the SAFE score to data from 7,156 participants of the National Health and Nutrition Examination Survey (NHANES) for 2017-2020. NHANES is representative of the noninstitutionalized, civilian population of the United States. It includes broad demographic, clinical, and laboratory data, including transient elastography data. FibroScans were first used in 2017, so the investigators had 3 years of FibroScan data with which to validate the score.

The researchers extrapolated the NHANES sample data to the U.S. population. They found that the proportion of adults with steatosis (CAP score > 274 dB/m) and significant fibrosis (LSM > 8.0 kPa) was 42.7% (95% confidence interval, 41.0%- 44.3%) and 8.9% (7.6%-10.2%), respectively. In addition, 11.3% (10.2%-12.5%) of the adult U.S. population demonstrated a significant amount of alcohol use, 2.3% (1.4%-3.3%) showed evidence of hepatitis B or C, and 5.4% (4.6%-6.2%) had elevated serum ferritin levels.

The researchers then stratified the patients according to previously defined SAFE tiers of low, intermediate, and high risk and projected findings to the U.S. general population.

“When we applied our score to the general population, we found multiple abnormalities in the high-risk groups [SAFE >100] having Fibroscan data that are consistent with stage 2 or higher fibrosis regardless of etiology, “Dr. Kim pointed out.

Results also showed that very few patients with SAFE less than 0 had liver fibrosis (4% among those with liver stiffness measure [LSM] > 8kPa, and 0.8% with LSM > 12kPa). Among those with SAFE > 100, nearly a third (31.5%) had LSM of > 8kPa, and 16.5% had LSM > 12kPa.

In addition to fibrosis, liver abnormalities were common among patients with SAFE greater than 100, including steatosis (68.0%), viral hepatitis (7.0%), and abnormal ferritin levels (12.9%); 10.8% of these patients used alcohol.

“Right now, some patients are referred, but on examination and FibroScan, they might actually be okay, so it it’s a waste of time and money for everyone. We can preempt all of this by doing a blood test and focusing on those people who really need a scan,” said Dr. Kim.

The researcher is now working with primary care colleagues to help further develop and integrate SAFE into the primary care setting.
 

Fibrosis score in patients with metabolic dysfunction

Also presenting at the same session on population health was Willem Pieter Brouwer, MD, PhD, from Erasmus University Medical Center, Rotterdam, the Netherlands. He reported results of a validation study of a new risk score – the Metabolic Dysfunction-Associated Fibrosis–5 (MAF-5) score – for use for people with metabolic dysfunction who are recommended for screening for liver fibrosis.

“We believe the MAF-5 score may be a good alternative to the FIB-4 [a liver fibrosis biomarker] for use in the referral pathway for liver health evaluation,” remarked Dr. Brouwer. “The clinical practice guidelines recommend using FIB-4 scores, but these have a poor-moderate performance in the population setting.

“We developed and validated our score in a population of 5,500 from the NHANES 2017-2020 cycle and validated the score in populations from Rotterdam, which is a cohort of elderly participants, and in fibrosis among patients with biopsy-proven NAFLD from Colombia and Belgium,” he explained.

He also validated the score against different existing scoring systems and different methods of measuring liver stiffness and validated it for prognostic use to predict all-cause mortality in the NHANES III cohort.

Dr. Brouwer removed age as a factor of his new MAF-5 score; the score is thus stable for patients of all ages and is suitable for detecting liver disease in younger patients. “This is very important because these patients are currently underserved and have the most years of life to win.”

Referring to the SAFE score discussed by Dr. Kim, as well as other scores, he said, “The FIB-4, SAFE, and NFS [NAFLD fibrosis score] all include age in the scores, which causes problems and limitations in aging populations, as more and more patients will be referred due to an increasing score. Hence, the elderly are mostly all referred for liver checkups.”

Dr. Kim and Dr. Brouwer have disclosed no relevant financial relationships.

A version of this article first appeared on Medscape.com.

VIENNA – An algorithm, the Steatosis-Associated Fibrosis Estimator (SAFE), was developed to detect clinically significant fibrosis in patients with nonalcoholic fatty liver disease (NAFLD). It is effective at detecting chronic liver disease from all causes with or without NAFLD in the general population, according the results of a U.S. population-based study. The algorithm was designed for use in primary care to help slow the steep rise in liver disease burden.

On the basis of the SAFE score, 61.3% of participants were at low risk for clinically significant fibrosis; 11.2% were at high risk; and 27.5% were at intermediate risk. Upon validation, very few of the low-risk participants had liver fibrosis, while nearly a third of those with a high-risk score had clinically significant fibrosis. In addition, a high percentage of the patients with high-risk SAFE scores had viral hepatitis and elevations in ferritin level.

“This is the first time that there has been a test that provides a score to detect low-risk liver disease in primary care,” said Ray Kim, MD, from Stanford (Calif.) University, senior investigator, who was speaking to this news organization at the annual International Liver Congress sponsored by the European Association for the Study of the Liver

“Primary care doctors currently detect liver disease through a serendipitous abnormal finding on ultrasound or blood tests that detect elevated transaminases, and then the patient is referred to a hepatologist, who figures out what is really going on,” said Dr. Kim.

“This approach is limited, so we need to get SAFE into primary care so these doctors can automatically calculate their scores, and if the patient is over 100 [high risk of chronic liver disease], then they need help [referral to a hepatologist].”
 

Liver deaths sharply rising

Public health data show that more people are dying of liver disease today than previously. Deaths in the United States have doubled over the past 20 years, said Dr. Kim. “If our mission is to help these patients and prevent death, [things are] moving in the wrong direction.”

He stressed that in order to change the direction, “primary care doctors need to engage with the issue and have appropriate tools to identify people with liver disease.”

Most often, the reason for this rise in deaths is that cases are being diagnosed at advanced stages of disease in which reversibility is limited, he added. “We want to move upstream where people might have early-stage disease and where we can intervene and make a difference.”

In an effort to help earlier detection of liver disease, the SAFE score was developed and validated by Dr. Kim and his colleagues to detect clinically significant (greater than stage 2) fibrosis in patients with NAFLD in primary care. The score is based upon age, body mass index, diabetes, platelet level, aspartate and alanine aminotransferase levels, and globulin level. A score of less than zero signifies that a patient is at low risk for liver fibrosis, while a score greater than 100 signifies a high risk of fibrosis. A score between 0 and 99 denotes intermediate risk of fibrosis.

“Unlike other noninvasive tests that detect advanced fibrosis, this one detects early-stage fibrosis. We’ve shown that the SAFE estimator is better than all other blood-based markers,” explained Dr. Kim.
 

 

 

Applying SAFE to the general population

In the study presented here at EASL, Dr. Kim aimed to expand the horizon for SAFE testing to the U.S. general population and to assess whether SAFE was effective in screening for chronic liver disease regardless of steatosis of the liver.

Together with first author Nakia Chung, MD, also from Stanford University, Dr. Kim applied the SAFE score to data from 7,156 participants of the National Health and Nutrition Examination Survey (NHANES) for 2017-2020. NHANES is representative of the noninstitutionalized, civilian population of the United States. It includes broad demographic, clinical, and laboratory data, including transient elastography data. FibroScans were first used in 2017, so the investigators had 3 years of FibroScan data with which to validate the score.

The researchers extrapolated the NHANES sample data to the U.S. population. They found that the proportion of adults with steatosis (CAP score > 274 dB/m) and significant fibrosis (LSM > 8.0 kPa) was 42.7% (95% confidence interval, 41.0%- 44.3%) and 8.9% (7.6%-10.2%), respectively. In addition, 11.3% (10.2%-12.5%) of the adult U.S. population demonstrated a significant amount of alcohol use, 2.3% (1.4%-3.3%) showed evidence of hepatitis B or C, and 5.4% (4.6%-6.2%) had elevated serum ferritin levels.

The researchers then stratified the patients according to previously defined SAFE tiers of low, intermediate, and high risk and projected findings to the U.S. general population.

“When we applied our score to the general population, we found multiple abnormalities in the high-risk groups [SAFE >100] having Fibroscan data that are consistent with stage 2 or higher fibrosis regardless of etiology, “Dr. Kim pointed out.

Results also showed that very few patients with SAFE less than 0 had liver fibrosis (4% among those with liver stiffness measure [LSM] > 8kPa, and 0.8% with LSM > 12kPa). Among those with SAFE > 100, nearly a third (31.5%) had LSM of > 8kPa, and 16.5% had LSM > 12kPa.

In addition to fibrosis, liver abnormalities were common among patients with SAFE greater than 100, including steatosis (68.0%), viral hepatitis (7.0%), and abnormal ferritin levels (12.9%); 10.8% of these patients used alcohol.

“Right now, some patients are referred, but on examination and FibroScan, they might actually be okay, so it it’s a waste of time and money for everyone. We can preempt all of this by doing a blood test and focusing on those people who really need a scan,” said Dr. Kim.

The researcher is now working with primary care colleagues to help further develop and integrate SAFE into the primary care setting.
 

Fibrosis score in patients with metabolic dysfunction

Also presenting at the same session on population health was Willem Pieter Brouwer, MD, PhD, from Erasmus University Medical Center, Rotterdam, the Netherlands. He reported results of a validation study of a new risk score – the Metabolic Dysfunction-Associated Fibrosis–5 (MAF-5) score – for use for people with metabolic dysfunction who are recommended for screening for liver fibrosis.

“We believe the MAF-5 score may be a good alternative to the FIB-4 [a liver fibrosis biomarker] for use in the referral pathway for liver health evaluation,” remarked Dr. Brouwer. “The clinical practice guidelines recommend using FIB-4 scores, but these have a poor-moderate performance in the population setting.

“We developed and validated our score in a population of 5,500 from the NHANES 2017-2020 cycle and validated the score in populations from Rotterdam, which is a cohort of elderly participants, and in fibrosis among patients with biopsy-proven NAFLD from Colombia and Belgium,” he explained.

He also validated the score against different existing scoring systems and different methods of measuring liver stiffness and validated it for prognostic use to predict all-cause mortality in the NHANES III cohort.

Dr. Brouwer removed age as a factor of his new MAF-5 score; the score is thus stable for patients of all ages and is suitable for detecting liver disease in younger patients. “This is very important because these patients are currently underserved and have the most years of life to win.”

Referring to the SAFE score discussed by Dr. Kim, as well as other scores, he said, “The FIB-4, SAFE, and NFS [NAFLD fibrosis score] all include age in the scores, which causes problems and limitations in aging populations, as more and more patients will be referred due to an increasing score. Hence, the elderly are mostly all referred for liver checkups.”

Dr. Kim and Dr. Brouwer have disclosed no relevant financial relationships.

A version of this article first appeared on Medscape.com.

Publications
Publications
Topics
Article Type
Sections
Article Source

AT ILC 2023

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article