LayerRx Mapping ID
952
Slot System
Featured Buckets
Featured Buckets Admin
Reverse Chronological Sort

Global project reveals cancer’s genomic playbook

Article Type
Changed
Thu, 12/15/2022 - 17:39

A massive collaborative project spanning four continents and 744 research centers has revealed driver mutations in both protein-coding and noncoding regions of 38 cancer types.

Pan-Cancer Analysis of Whole Genomes
The Pan-Cancer Analysis of Whole Genomes Consortium analyzed more than 2,600 tumor samples from patients with 38 cancer types.

The Pan-Cancer Analysis of Whole Genomes (PCAWG) is an integrative analysis of the whole-genome sequences from 2,658 donors across 38 common tumor types. The findings are expected to add exponentially to what’s currently known about the complex genetics of cancer, and they point to possible strategies for improving cancer prevention, diagnosis, and care.

Six articles summarizing the findings are presented in a series of papers in Nature, and 16 more appear in affiliated publications.

“It’s humbling that it was only 14 years ago that the genomics community sequenced its very first cancer exome, and it was able to identify mutations within the roughly 20,000 protein-coding genes in the human cell,” investigator Lincoln Stein, MD, PhD, of the Ontario Institute for Cancer Research in Toronto, said in a telephone briefing.

Exome sequencing, however, covers only protein-coding genomic regions, which constitute only about 1% of the entire genome, “so assembling an accurate portrait of the cancer genome using just the exome data is like trying to put together a 100,000-piece jigsaw puzzle when you’re missing 99% of the pieces and there’s no puzzle box with a completed picture to guide you,” Dr. Stein said.

Members of the PCAWG from centers in North America, Europe, Asia, and Australia screened 2,658 whole-cancer genomes and matched samples of noncancerous tissues from the same individuals, along with 1,188 transcriptomes cataloging the sequences and expression of RNA transcripts in a given tumor. The 6-year project netted more than 800 terabytes of genomic data, roughly equivalent to the digital holdings of the U.S. Library of Congress multiplied by 11.

The findings are summarized in papers focusing on cancer drivers, noncoding changes, mutational signatures, structural variants, cancer evolution over time, and RNA alterations.
 

Driver mutations

Investigators found that the average cancer genome contains four or five driver mutations located in both coding and noncoding regions. They also found, however, that in approximately 5% of cases no driver mutations could be identified.

A substantial proportion of tumors displayed “hallmarks of genomic catastrophes.” About 22% of tumors exhibited chromothripsis, a mutational process marked by hundreds or even thousands of clustered chromosomal rearrangements. About 18% showed chromoplexy, which is characterized by scattering and rearrangement of multiple strands of DNA from one or more chromosomes.

Analyzing driver point mutations and structural variants in noncoding regions, the investigators found the usual suspects – previously reported culprits – as well as novel candidates.

For example, they identified point mutations in the five prime region of the tumor suppressor gene TP53 and the three prime untranslated regions of NFKBIZ (a nuclear factor kappa B inhibitor) and TOB1 (an antiproliferative protein), focal deletion in BRD4 (a transcriptional and epigenetic regulator), and rearrangements in chromosomal loci in members of the AKR1C family of enzymes thought to play a role in disease progression.

In addition, investigators identified mutations in noncoding regions of TERT, a telomerase gene. These mutations result in ramped-up expression of telomerase, which in turn promotes uncontrollable division of tumor cells.
 

 

 

Mutational signatures

In a related line of research, PCAWG investigators identified new DNA mutational signatures ranging from single nucleotide polymorphisms to insertions and deletions, as well as to structural variants – rearrangements of large sections of the genome.

“The substantial size of our dataset, compared with previous analyses, enabled the discovery of new signatures, the separation of overlapping signatures, and the decomposition of signatures into components that may represent associated – but distinct – DNA damage, repair, and/or replication mechanisms. By estimating the contribution of each signature to the mutational catalogs of individual cancer genomes, we revealed associations of signatures to exogenous or endogenous exposures, as well as to defective DNA maintenance processes,” the investigators wrote.

They also acknowledged, however, that “many signatures are of unknown cause.”
 

Cancer evolution

One of the six main studies focused on the evolution of cancer over time. Instead of providing a “snapshot” of the genome as captured by sequencing tissue from a single biopsy, consortium investigators created full-length features of the “life history and evolution of mutational processes and driver mutation sequences.”

They found that early cancer development was marked by relatively few mutations in driver genes and by identifiable copy-number gains, including trisomy 7 in glioblastoma, and an abnormal mirroring of the arms (isochromosome) of chromosome 17 in medulloblastoma.

In 40% of the samples, however, there were significant changes in the mutational spectrum as the cancers grew, leading to a near quadrupling of driver genes and increased genomic instability in later-stage tumors.

“Copy-number alterations often occur in mitotic crises and lead to simultaneous gains of chromosomal segments,” the investigators wrote. “Timing analyses suggest that driver mutations often precede diagnosis by many years, if not decades. Together, these results determine the evolutionary trajectories of cancer and highlight opportunities for early cancer detection.”
 

Implications for cancer care

“When I used to treat patients with cancer, I was always completely amazed and puzzled by how two patients could have what looked like the same tumor. It would look the same under the microscope, have the same size, and the two patients would receive exactly the same treatment, but the two patients would have completely opposite outcomes; one would survive, and one would die. What this analysis … has done is really laid bare the reasons for that unpredictability in clinical outcomes,” Peter Campbell, MD, PhD, of the Wellcome Sanger Institute in Hinxton, England, said during the telebriefing.

“The most striking finding out of all of the suite of papers is just how different one person’s cancer genome is from another person’s. We see thousands of different combinations of mutations that can cause the cancer, and more than 80 different underlying processes generating the mutations in a cancer, and that leads to very different shapes and patterns in the genome that result,” he added.

On a positive note, the research shows that one or more driver mutations can be identified in about 95% of all cancer patients, and it elucidates the sequence of events leading to oncogenesis and tumor evolution, providing opportunities for earlier identification and potential interventions to prevent cancer, Dr. Campbell said.

The PCAWG was a collaborative multinational effort with multiple funding sources and many investigators.

SOURCE: Nature. 2020 Feb 5. https://www.nature.com/collections/pcawg/

Publications
Topics
Sections

A massive collaborative project spanning four continents and 744 research centers has revealed driver mutations in both protein-coding and noncoding regions of 38 cancer types.

Pan-Cancer Analysis of Whole Genomes
The Pan-Cancer Analysis of Whole Genomes Consortium analyzed more than 2,600 tumor samples from patients with 38 cancer types.

The Pan-Cancer Analysis of Whole Genomes (PCAWG) is an integrative analysis of the whole-genome sequences from 2,658 donors across 38 common tumor types. The findings are expected to add exponentially to what’s currently known about the complex genetics of cancer, and they point to possible strategies for improving cancer prevention, diagnosis, and care.

Six articles summarizing the findings are presented in a series of papers in Nature, and 16 more appear in affiliated publications.

“It’s humbling that it was only 14 years ago that the genomics community sequenced its very first cancer exome, and it was able to identify mutations within the roughly 20,000 protein-coding genes in the human cell,” investigator Lincoln Stein, MD, PhD, of the Ontario Institute for Cancer Research in Toronto, said in a telephone briefing.

Exome sequencing, however, covers only protein-coding genomic regions, which constitute only about 1% of the entire genome, “so assembling an accurate portrait of the cancer genome using just the exome data is like trying to put together a 100,000-piece jigsaw puzzle when you’re missing 99% of the pieces and there’s no puzzle box with a completed picture to guide you,” Dr. Stein said.

Members of the PCAWG from centers in North America, Europe, Asia, and Australia screened 2,658 whole-cancer genomes and matched samples of noncancerous tissues from the same individuals, along with 1,188 transcriptomes cataloging the sequences and expression of RNA transcripts in a given tumor. The 6-year project netted more than 800 terabytes of genomic data, roughly equivalent to the digital holdings of the U.S. Library of Congress multiplied by 11.

The findings are summarized in papers focusing on cancer drivers, noncoding changes, mutational signatures, structural variants, cancer evolution over time, and RNA alterations.
 

Driver mutations

Investigators found that the average cancer genome contains four or five driver mutations located in both coding and noncoding regions. They also found, however, that in approximately 5% of cases no driver mutations could be identified.

A substantial proportion of tumors displayed “hallmarks of genomic catastrophes.” About 22% of tumors exhibited chromothripsis, a mutational process marked by hundreds or even thousands of clustered chromosomal rearrangements. About 18% showed chromoplexy, which is characterized by scattering and rearrangement of multiple strands of DNA from one or more chromosomes.

Analyzing driver point mutations and structural variants in noncoding regions, the investigators found the usual suspects – previously reported culprits – as well as novel candidates.

For example, they identified point mutations in the five prime region of the tumor suppressor gene TP53 and the three prime untranslated regions of NFKBIZ (a nuclear factor kappa B inhibitor) and TOB1 (an antiproliferative protein), focal deletion in BRD4 (a transcriptional and epigenetic regulator), and rearrangements in chromosomal loci in members of the AKR1C family of enzymes thought to play a role in disease progression.

In addition, investigators identified mutations in noncoding regions of TERT, a telomerase gene. These mutations result in ramped-up expression of telomerase, which in turn promotes uncontrollable division of tumor cells.
 

 

 

Mutational signatures

In a related line of research, PCAWG investigators identified new DNA mutational signatures ranging from single nucleotide polymorphisms to insertions and deletions, as well as to structural variants – rearrangements of large sections of the genome.

“The substantial size of our dataset, compared with previous analyses, enabled the discovery of new signatures, the separation of overlapping signatures, and the decomposition of signatures into components that may represent associated – but distinct – DNA damage, repair, and/or replication mechanisms. By estimating the contribution of each signature to the mutational catalogs of individual cancer genomes, we revealed associations of signatures to exogenous or endogenous exposures, as well as to defective DNA maintenance processes,” the investigators wrote.

They also acknowledged, however, that “many signatures are of unknown cause.”
 

Cancer evolution

One of the six main studies focused on the evolution of cancer over time. Instead of providing a “snapshot” of the genome as captured by sequencing tissue from a single biopsy, consortium investigators created full-length features of the “life history and evolution of mutational processes and driver mutation sequences.”

They found that early cancer development was marked by relatively few mutations in driver genes and by identifiable copy-number gains, including trisomy 7 in glioblastoma, and an abnormal mirroring of the arms (isochromosome) of chromosome 17 in medulloblastoma.

In 40% of the samples, however, there were significant changes in the mutational spectrum as the cancers grew, leading to a near quadrupling of driver genes and increased genomic instability in later-stage tumors.

“Copy-number alterations often occur in mitotic crises and lead to simultaneous gains of chromosomal segments,” the investigators wrote. “Timing analyses suggest that driver mutations often precede diagnosis by many years, if not decades. Together, these results determine the evolutionary trajectories of cancer and highlight opportunities for early cancer detection.”
 

Implications for cancer care

“When I used to treat patients with cancer, I was always completely amazed and puzzled by how two patients could have what looked like the same tumor. It would look the same under the microscope, have the same size, and the two patients would receive exactly the same treatment, but the two patients would have completely opposite outcomes; one would survive, and one would die. What this analysis … has done is really laid bare the reasons for that unpredictability in clinical outcomes,” Peter Campbell, MD, PhD, of the Wellcome Sanger Institute in Hinxton, England, said during the telebriefing.

“The most striking finding out of all of the suite of papers is just how different one person’s cancer genome is from another person’s. We see thousands of different combinations of mutations that can cause the cancer, and more than 80 different underlying processes generating the mutations in a cancer, and that leads to very different shapes and patterns in the genome that result,” he added.

On a positive note, the research shows that one or more driver mutations can be identified in about 95% of all cancer patients, and it elucidates the sequence of events leading to oncogenesis and tumor evolution, providing opportunities for earlier identification and potential interventions to prevent cancer, Dr. Campbell said.

The PCAWG was a collaborative multinational effort with multiple funding sources and many investigators.

SOURCE: Nature. 2020 Feb 5. https://www.nature.com/collections/pcawg/

A massive collaborative project spanning four continents and 744 research centers has revealed driver mutations in both protein-coding and noncoding regions of 38 cancer types.

Pan-Cancer Analysis of Whole Genomes
The Pan-Cancer Analysis of Whole Genomes Consortium analyzed more than 2,600 tumor samples from patients with 38 cancer types.

The Pan-Cancer Analysis of Whole Genomes (PCAWG) is an integrative analysis of the whole-genome sequences from 2,658 donors across 38 common tumor types. The findings are expected to add exponentially to what’s currently known about the complex genetics of cancer, and they point to possible strategies for improving cancer prevention, diagnosis, and care.

Six articles summarizing the findings are presented in a series of papers in Nature, and 16 more appear in affiliated publications.

“It’s humbling that it was only 14 years ago that the genomics community sequenced its very first cancer exome, and it was able to identify mutations within the roughly 20,000 protein-coding genes in the human cell,” investigator Lincoln Stein, MD, PhD, of the Ontario Institute for Cancer Research in Toronto, said in a telephone briefing.

Exome sequencing, however, covers only protein-coding genomic regions, which constitute only about 1% of the entire genome, “so assembling an accurate portrait of the cancer genome using just the exome data is like trying to put together a 100,000-piece jigsaw puzzle when you’re missing 99% of the pieces and there’s no puzzle box with a completed picture to guide you,” Dr. Stein said.

Members of the PCAWG from centers in North America, Europe, Asia, and Australia screened 2,658 whole-cancer genomes and matched samples of noncancerous tissues from the same individuals, along with 1,188 transcriptomes cataloging the sequences and expression of RNA transcripts in a given tumor. The 6-year project netted more than 800 terabytes of genomic data, roughly equivalent to the digital holdings of the U.S. Library of Congress multiplied by 11.

The findings are summarized in papers focusing on cancer drivers, noncoding changes, mutational signatures, structural variants, cancer evolution over time, and RNA alterations.
 

Driver mutations

Investigators found that the average cancer genome contains four or five driver mutations located in both coding and noncoding regions. They also found, however, that in approximately 5% of cases no driver mutations could be identified.

A substantial proportion of tumors displayed “hallmarks of genomic catastrophes.” About 22% of tumors exhibited chromothripsis, a mutational process marked by hundreds or even thousands of clustered chromosomal rearrangements. About 18% showed chromoplexy, which is characterized by scattering and rearrangement of multiple strands of DNA from one or more chromosomes.

Analyzing driver point mutations and structural variants in noncoding regions, the investigators found the usual suspects – previously reported culprits – as well as novel candidates.

For example, they identified point mutations in the five prime region of the tumor suppressor gene TP53 and the three prime untranslated regions of NFKBIZ (a nuclear factor kappa B inhibitor) and TOB1 (an antiproliferative protein), focal deletion in BRD4 (a transcriptional and epigenetic regulator), and rearrangements in chromosomal loci in members of the AKR1C family of enzymes thought to play a role in disease progression.

In addition, investigators identified mutations in noncoding regions of TERT, a telomerase gene. These mutations result in ramped-up expression of telomerase, which in turn promotes uncontrollable division of tumor cells.
 

 

 

Mutational signatures

In a related line of research, PCAWG investigators identified new DNA mutational signatures ranging from single nucleotide polymorphisms to insertions and deletions, as well as to structural variants – rearrangements of large sections of the genome.

“The substantial size of our dataset, compared with previous analyses, enabled the discovery of new signatures, the separation of overlapping signatures, and the decomposition of signatures into components that may represent associated – but distinct – DNA damage, repair, and/or replication mechanisms. By estimating the contribution of each signature to the mutational catalogs of individual cancer genomes, we revealed associations of signatures to exogenous or endogenous exposures, as well as to defective DNA maintenance processes,” the investigators wrote.

They also acknowledged, however, that “many signatures are of unknown cause.”
 

Cancer evolution

One of the six main studies focused on the evolution of cancer over time. Instead of providing a “snapshot” of the genome as captured by sequencing tissue from a single biopsy, consortium investigators created full-length features of the “life history and evolution of mutational processes and driver mutation sequences.”

They found that early cancer development was marked by relatively few mutations in driver genes and by identifiable copy-number gains, including trisomy 7 in glioblastoma, and an abnormal mirroring of the arms (isochromosome) of chromosome 17 in medulloblastoma.

In 40% of the samples, however, there were significant changes in the mutational spectrum as the cancers grew, leading to a near quadrupling of driver genes and increased genomic instability in later-stage tumors.

“Copy-number alterations often occur in mitotic crises and lead to simultaneous gains of chromosomal segments,” the investigators wrote. “Timing analyses suggest that driver mutations often precede diagnosis by many years, if not decades. Together, these results determine the evolutionary trajectories of cancer and highlight opportunities for early cancer detection.”
 

Implications for cancer care

“When I used to treat patients with cancer, I was always completely amazed and puzzled by how two patients could have what looked like the same tumor. It would look the same under the microscope, have the same size, and the two patients would receive exactly the same treatment, but the two patients would have completely opposite outcomes; one would survive, and one would die. What this analysis … has done is really laid bare the reasons for that unpredictability in clinical outcomes,” Peter Campbell, MD, PhD, of the Wellcome Sanger Institute in Hinxton, England, said during the telebriefing.

“The most striking finding out of all of the suite of papers is just how different one person’s cancer genome is from another person’s. We see thousands of different combinations of mutations that can cause the cancer, and more than 80 different underlying processes generating the mutations in a cancer, and that leads to very different shapes and patterns in the genome that result,” he added.

On a positive note, the research shows that one or more driver mutations can be identified in about 95% of all cancer patients, and it elucidates the sequence of events leading to oncogenesis and tumor evolution, providing opportunities for earlier identification and potential interventions to prevent cancer, Dr. Campbell said.

The PCAWG was a collaborative multinational effort with multiple funding sources and many investigators.

SOURCE: Nature. 2020 Feb 5. https://www.nature.com/collections/pcawg/

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM NATURE

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.

Preoperative CT shows little value in early vulvar SCC

Article Type
Changed
Tue, 02/04/2020 - 12:57

Preoperative computerized tomography (CT) demonstrated limited value in the clinical management of patients with early-stage vulvar squamous cell carcinoma (VSCC) analyzed in a single-center study.

The findings suggest preoperative CT imaging could be excluded prior to sentinel inguinal lymph node biopsy or staging surgery in patients with early-stage disease.

“In this study, we aimed to investigate if preoperative CT scan influences the overall course of VSCC management in patients without clinical evidence of groin lymphadenopathy,” wrote Rachel Pounds, MD, of the University of Birmingham (England) and colleagues. The study was published in Gynecologic Oncology.

The researchers prospectively studied a cohort of 225 patients with primary or recurrent VSCC who underwent staging surgery at a single institution in the United Kingdom. The patients’ mean age was 67 years (range, 54-79 years), and most had stage 1B disease (57.8%).

The researchers compared preoperative imaging findings with histological results from sentinel inguinal lymph node biopsy. Other clinical information, including surgery type, evidence of groin node involvement, and age at diagnosis was collected from patient files and included in the analysis.

In all, 51.6% of patients underwent preoperative CT imaging. Among these patients, 37.9% had a positive report of radiological groin lymphatic metastases.

“True groin node metastases, confirmed histologically, were observed in 26 patients (22.4%) with a radiologically positive scan report (true positives) and in 18 patients (15.5%) with a radiological negative scan report (false negatives),” the researchers wrote.

The specificity and sensitivity of preoperative CT to detect groin lymphatic metastasis were 77.8% and 59.1%, respectively. The positive and negative predictive values were 61.9% and 75.7%, respectively.

There was no significant difference in overall survival, disease-specific or disease-free survival, or groin node recurrence between patients who underwent preoperative CT and patients who did not.

Groin node recurrence was observed in 10.3% of patients with preoperative CT and 11.5% of patients without it (P = .7768). Disease-specific death occurred in 16.4% of patients with preoperative CT and 13.5% of patients without it (P = .5451).

“Our results highlight the poor reliability of preoperative CT scans in detecting inguinal lymphatic metastasis,” the researchers wrote. “Preoperative CT scan may be omitted in early stage VSCC prior to surgical staging as it does not affect overall management and surgical outcomes.”

No funding sources were reported for this study. The authors reported having no conflicts of interest.

SOURCE: Pounds R et al. Gynecol Oncol. 2020 Jan 24. doi: 10.1016/j.ygyno.2020.01.031.

Publications
Topics
Sections

Preoperative computerized tomography (CT) demonstrated limited value in the clinical management of patients with early-stage vulvar squamous cell carcinoma (VSCC) analyzed in a single-center study.

The findings suggest preoperative CT imaging could be excluded prior to sentinel inguinal lymph node biopsy or staging surgery in patients with early-stage disease.

“In this study, we aimed to investigate if preoperative CT scan influences the overall course of VSCC management in patients without clinical evidence of groin lymphadenopathy,” wrote Rachel Pounds, MD, of the University of Birmingham (England) and colleagues. The study was published in Gynecologic Oncology.

The researchers prospectively studied a cohort of 225 patients with primary or recurrent VSCC who underwent staging surgery at a single institution in the United Kingdom. The patients’ mean age was 67 years (range, 54-79 years), and most had stage 1B disease (57.8%).

The researchers compared preoperative imaging findings with histological results from sentinel inguinal lymph node biopsy. Other clinical information, including surgery type, evidence of groin node involvement, and age at diagnosis was collected from patient files and included in the analysis.

In all, 51.6% of patients underwent preoperative CT imaging. Among these patients, 37.9% had a positive report of radiological groin lymphatic metastases.

“True groin node metastases, confirmed histologically, were observed in 26 patients (22.4%) with a radiologically positive scan report (true positives) and in 18 patients (15.5%) with a radiological negative scan report (false negatives),” the researchers wrote.

The specificity and sensitivity of preoperative CT to detect groin lymphatic metastasis were 77.8% and 59.1%, respectively. The positive and negative predictive values were 61.9% and 75.7%, respectively.

There was no significant difference in overall survival, disease-specific or disease-free survival, or groin node recurrence between patients who underwent preoperative CT and patients who did not.

Groin node recurrence was observed in 10.3% of patients with preoperative CT and 11.5% of patients without it (P = .7768). Disease-specific death occurred in 16.4% of patients with preoperative CT and 13.5% of patients without it (P = .5451).

“Our results highlight the poor reliability of preoperative CT scans in detecting inguinal lymphatic metastasis,” the researchers wrote. “Preoperative CT scan may be omitted in early stage VSCC prior to surgical staging as it does not affect overall management and surgical outcomes.”

No funding sources were reported for this study. The authors reported having no conflicts of interest.

SOURCE: Pounds R et al. Gynecol Oncol. 2020 Jan 24. doi: 10.1016/j.ygyno.2020.01.031.

Preoperative computerized tomography (CT) demonstrated limited value in the clinical management of patients with early-stage vulvar squamous cell carcinoma (VSCC) analyzed in a single-center study.

The findings suggest preoperative CT imaging could be excluded prior to sentinel inguinal lymph node biopsy or staging surgery in patients with early-stage disease.

“In this study, we aimed to investigate if preoperative CT scan influences the overall course of VSCC management in patients without clinical evidence of groin lymphadenopathy,” wrote Rachel Pounds, MD, of the University of Birmingham (England) and colleagues. The study was published in Gynecologic Oncology.

The researchers prospectively studied a cohort of 225 patients with primary or recurrent VSCC who underwent staging surgery at a single institution in the United Kingdom. The patients’ mean age was 67 years (range, 54-79 years), and most had stage 1B disease (57.8%).

The researchers compared preoperative imaging findings with histological results from sentinel inguinal lymph node biopsy. Other clinical information, including surgery type, evidence of groin node involvement, and age at diagnosis was collected from patient files and included in the analysis.

In all, 51.6% of patients underwent preoperative CT imaging. Among these patients, 37.9% had a positive report of radiological groin lymphatic metastases.

“True groin node metastases, confirmed histologically, were observed in 26 patients (22.4%) with a radiologically positive scan report (true positives) and in 18 patients (15.5%) with a radiological negative scan report (false negatives),” the researchers wrote.

The specificity and sensitivity of preoperative CT to detect groin lymphatic metastasis were 77.8% and 59.1%, respectively. The positive and negative predictive values were 61.9% and 75.7%, respectively.

There was no significant difference in overall survival, disease-specific or disease-free survival, or groin node recurrence between patients who underwent preoperative CT and patients who did not.

Groin node recurrence was observed in 10.3% of patients with preoperative CT and 11.5% of patients without it (P = .7768). Disease-specific death occurred in 16.4% of patients with preoperative CT and 13.5% of patients without it (P = .5451).

“Our results highlight the poor reliability of preoperative CT scans in detecting inguinal lymphatic metastasis,” the researchers wrote. “Preoperative CT scan may be omitted in early stage VSCC prior to surgical staging as it does not affect overall management and surgical outcomes.”

No funding sources were reported for this study. The authors reported having no conflicts of interest.

SOURCE: Pounds R et al. Gynecol Oncol. 2020 Jan 24. doi: 10.1016/j.ygyno.2020.01.031.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM GYNECOLOGIC ONCOLOGY

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.

Ovarian cancer survival varies between high-income countries

Article Type
Changed
Tue, 02/04/2020 - 15:08

Considerable differences in ovarian cancer survival exist between high-income countries, especially for older women with advanced disease, according to a recent study.

The findings suggest additional research is needed to evaluate international differences in both treatment- and patient-specific factors affecting survival.

“This study [evaluated] differences in survival by age and stage at diagnosis within and across seven high-income countries,” wrote Citadel J. Cabasag, PhD, of the International Agency for Research on Cancer in Lyon, France, and colleagues. The results were published in Gynecologic Oncology.

The researchers conducted a retrospective analysis of population-based registry data from 2010 to 2014. Data were collected from 21 cancer registries located in Canada, United Kingdom, New Zealand, Norway, Ireland, Australia, and Denmark.

The cohort included 58,161 women with epithelial and nonepithelial ovarian cancer. The majority of cases were advanced stage, with a median age of 63-67 years at diagnosis, depending on the country.

The researchers compared age- and stage-specific net survival between countries at 1 and 3 years post diagnosis.

The 3-year all-ages net survival was highest for Norway (57%) and Australia (56%), followed by Denmark (54%), Canada (50%), United Kingdom (47%), New Zealand (46%), and Ireland (45%).

Most patients were diagnosed with distant disease (range, 64%-71%), with the greatest proportion of women in the 65- to 74- and 75- to 99-year age categories. The lowest 3-year age-specific survival (range, 20%-34%) was observed in the 75- to 99-year age category.

Marked differences in 3-year net survival between countries were found for women in the 75- to 99-year age group with distant disease (range, 12%-25%).

“International survival differences by age groups were less stark for early-stage disease,” the researchers reported. “Interjurisdictional differences within countries were also observed.”

The researchers acknowledged a key limitation of the analysis was the use of registry data. Variability between, and incomplete data within, registries could have lowered the accuracy of the survival estimates.

“[F]urther research investigating international differences in access to and quality of treatment, and prevalence of comorbid conditions particularly in older women with advanced disease [is needed],” the researchers concluded.

The study was supported by funding provided to the International Cancer Benchmarking Partnership. The authors reported having no conflicts of interest.

SOURCE: Cabasag CJ et al. Gynecol Oncol. 2020 Jan 28. doi: 10.1016/j.ygyno.2019.12.047.

Publications
Topics
Sections

Considerable differences in ovarian cancer survival exist between high-income countries, especially for older women with advanced disease, according to a recent study.

The findings suggest additional research is needed to evaluate international differences in both treatment- and patient-specific factors affecting survival.

“This study [evaluated] differences in survival by age and stage at diagnosis within and across seven high-income countries,” wrote Citadel J. Cabasag, PhD, of the International Agency for Research on Cancer in Lyon, France, and colleagues. The results were published in Gynecologic Oncology.

The researchers conducted a retrospective analysis of population-based registry data from 2010 to 2014. Data were collected from 21 cancer registries located in Canada, United Kingdom, New Zealand, Norway, Ireland, Australia, and Denmark.

The cohort included 58,161 women with epithelial and nonepithelial ovarian cancer. The majority of cases were advanced stage, with a median age of 63-67 years at diagnosis, depending on the country.

The researchers compared age- and stage-specific net survival between countries at 1 and 3 years post diagnosis.

The 3-year all-ages net survival was highest for Norway (57%) and Australia (56%), followed by Denmark (54%), Canada (50%), United Kingdom (47%), New Zealand (46%), and Ireland (45%).

Most patients were diagnosed with distant disease (range, 64%-71%), with the greatest proportion of women in the 65- to 74- and 75- to 99-year age categories. The lowest 3-year age-specific survival (range, 20%-34%) was observed in the 75- to 99-year age category.

Marked differences in 3-year net survival between countries were found for women in the 75- to 99-year age group with distant disease (range, 12%-25%).

“International survival differences by age groups were less stark for early-stage disease,” the researchers reported. “Interjurisdictional differences within countries were also observed.”

The researchers acknowledged a key limitation of the analysis was the use of registry data. Variability between, and incomplete data within, registries could have lowered the accuracy of the survival estimates.

“[F]urther research investigating international differences in access to and quality of treatment, and prevalence of comorbid conditions particularly in older women with advanced disease [is needed],” the researchers concluded.

The study was supported by funding provided to the International Cancer Benchmarking Partnership. The authors reported having no conflicts of interest.

SOURCE: Cabasag CJ et al. Gynecol Oncol. 2020 Jan 28. doi: 10.1016/j.ygyno.2019.12.047.

Considerable differences in ovarian cancer survival exist between high-income countries, especially for older women with advanced disease, according to a recent study.

The findings suggest additional research is needed to evaluate international differences in both treatment- and patient-specific factors affecting survival.

“This study [evaluated] differences in survival by age and stage at diagnosis within and across seven high-income countries,” wrote Citadel J. Cabasag, PhD, of the International Agency for Research on Cancer in Lyon, France, and colleagues. The results were published in Gynecologic Oncology.

The researchers conducted a retrospective analysis of population-based registry data from 2010 to 2014. Data were collected from 21 cancer registries located in Canada, United Kingdom, New Zealand, Norway, Ireland, Australia, and Denmark.

The cohort included 58,161 women with epithelial and nonepithelial ovarian cancer. The majority of cases were advanced stage, with a median age of 63-67 years at diagnosis, depending on the country.

The researchers compared age- and stage-specific net survival between countries at 1 and 3 years post diagnosis.

The 3-year all-ages net survival was highest for Norway (57%) and Australia (56%), followed by Denmark (54%), Canada (50%), United Kingdom (47%), New Zealand (46%), and Ireland (45%).

Most patients were diagnosed with distant disease (range, 64%-71%), with the greatest proportion of women in the 65- to 74- and 75- to 99-year age categories. The lowest 3-year age-specific survival (range, 20%-34%) was observed in the 75- to 99-year age category.

Marked differences in 3-year net survival between countries were found for women in the 75- to 99-year age group with distant disease (range, 12%-25%).

“International survival differences by age groups were less stark for early-stage disease,” the researchers reported. “Interjurisdictional differences within countries were also observed.”

The researchers acknowledged a key limitation of the analysis was the use of registry data. Variability between, and incomplete data within, registries could have lowered the accuracy of the survival estimates.

“[F]urther research investigating international differences in access to and quality of treatment, and prevalence of comorbid conditions particularly in older women with advanced disease [is needed],” the researchers concluded.

The study was supported by funding provided to the International Cancer Benchmarking Partnership. The authors reported having no conflicts of interest.

SOURCE: Cabasag CJ et al. Gynecol Oncol. 2020 Jan 28. doi: 10.1016/j.ygyno.2019.12.047.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM GYNECOLOGIC ONCOLOGY

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.

Lenvatinib/pembrolizumab has good activity in advanced RCC, other solid tumors

Article Type
Changed
Fri, 01/31/2020 - 14:02

 

A combination of the tyrosine kinase inhibitor lenvatinib (Lenvima) and the immune checkpoint inhibitor pembrolizumab (Keytruda) was safe and showed promising activity against advanced renal cell carcinoma and other solid tumors in a phase 1b/2 study.

Overall response rates (ORR) at 24 weeks ranged from 63% for patients with advanced renal cell carcinomas (RCC) to 25% for patients with urothelial cancers, reported Matthew H. Taylor, MD, of Knight Cancer Institute at Oregon Health & Science University in Portland, and colleagues.

The findings from this study sparked additional clinical trials for patients with gastric cancer, gastroesophageal cancer, and differentiated thyroid cancer, and set the stage for larger phase 3 trials in patients with advanced RCC, endometrial cancer, malignant melanoma, and non–small cell lung cancer (NSCLC).

“In the future, we also plan to study lenvatinib plus pembrolizumab in patients with RCC who have had disease progression after treatment with immune checkpoint inhibitors,” they wrote. The report was published in Journal of Clinical Oncology.

Lenvatinib is a multitargeted tyrosine kinase inhibitor (TKI) with action against vascular endothelial growth factor (VEGF) receptors 1-3, fibroblast growth factor (FGF) receptors 1-4, platelet-derived growth factor receptors alpha and the RET and KIT kinases.

“Preclinical and clinical studies suggest that modulation of VEGF-mediated immune suppression via angiogenesis inhibition could potentially augment the immunotherapeutic activity of immune checkpoint inhibitors,” the investigators wrote.

They reported results from the dose finding (1b) phase including 13 patients and initial phase 2 expansion cohorts with a total of 124 patients.

The maximum tolerated dose of lenvatinib in combination with pembrolizumab was established as 20 mg/day.

At 24 weeks of follow-up, the ORR for 30 patients with RCC was 63%; two additional patients had responses after week 24, for a total ORR at study cutoff in this cohort of 70%. The median duration of response for these patients was 20 months, and the median progression-free survival (PFS) was 19.8 months. At the time of data cutoff for this analysis, 9 of the 30 patients with RCC were still on treatment.

For 23 patients with endometrial cancer, the 24-week and overall ORR were 52%, with a median duration of response not reached, and a median PFS of 9.7 months. Seven patients were still on treatment at data cutoff.

For 21 patients with melanoma, the 24-week and overall ORR were 48%, median duration of response was 12.5 months, and median PFS was 5.5 months. Two of the patients were still on treatment at data cutoff.

For the 22 patients with squamous cell cancer of the head and neck, the 24-week ORR was 36%, with two patients having a response after week 24 for a total ORR at data cutoff of 46%. The median duration of response was 8.2 months and the median PFS was 4.7 months. Three patients remained on treatment at data cutoff.

For 21 patients with NSCLC, the 24-week and overall ORR were 33%, the median duration of response was 10.9 months, and median PFS was 5.9 months. Six of the patients were still receiving treatment at data cutoff.

For 20 patients with urothelial cancer, the 24-week and overall ORR were 25%, with a median duration of response not reached, and a median PFS of 5.4 months. Three patients were still receiving the combination at the time of data cutoff.

Treatment related adverse events (TRAEs) occurred in 133 of all 137 patients enrolled in the two study phases. The adverse events were similar across all cohorts, with any grade of events including fatigue in 58%, diarrhea in 52%, hypertension in 47%, hypothyroidism in 42%, and decreased appetite in 39%.

The most frequent grade 3 or 4 TRAEs were hypertension in 20%, fatigue in 12%, diarrhea in 9%, proteinuria in 8%, and increased lipase levels in 7%.

In all, 85% of patients had a TRAE leading to lenvatinib dose reduction and/or interruption, and 13% required lenvatinib discontinuation.

Events leading to pembrolizumab dose interruption occurred in 45% of patients, and pembrolizumab discontinuation in 15%.

The study was sponsored by Eisai with collaboration from Merck Sharp & Dohme. Dr. Taylor disclosed a consulting or advisory role for Bristol-Myers Squibb, Eisai, Array BioPharma, Loxo, Bayer, ArQule, Blueprint Medicines, Novartis, and Sanofi/Genzyme, and speakers bureau activities for BMS and Eisai.

SOURCE: Taylor MH et al. J Clin Oncol. 2020 Jan. 21 doi: 10.1200/JCO.19.01598.

Publications
Topics
Sections

 

A combination of the tyrosine kinase inhibitor lenvatinib (Lenvima) and the immune checkpoint inhibitor pembrolizumab (Keytruda) was safe and showed promising activity against advanced renal cell carcinoma and other solid tumors in a phase 1b/2 study.

Overall response rates (ORR) at 24 weeks ranged from 63% for patients with advanced renal cell carcinomas (RCC) to 25% for patients with urothelial cancers, reported Matthew H. Taylor, MD, of Knight Cancer Institute at Oregon Health & Science University in Portland, and colleagues.

The findings from this study sparked additional clinical trials for patients with gastric cancer, gastroesophageal cancer, and differentiated thyroid cancer, and set the stage for larger phase 3 trials in patients with advanced RCC, endometrial cancer, malignant melanoma, and non–small cell lung cancer (NSCLC).

“In the future, we also plan to study lenvatinib plus pembrolizumab in patients with RCC who have had disease progression after treatment with immune checkpoint inhibitors,” they wrote. The report was published in Journal of Clinical Oncology.

Lenvatinib is a multitargeted tyrosine kinase inhibitor (TKI) with action against vascular endothelial growth factor (VEGF) receptors 1-3, fibroblast growth factor (FGF) receptors 1-4, platelet-derived growth factor receptors alpha and the RET and KIT kinases.

“Preclinical and clinical studies suggest that modulation of VEGF-mediated immune suppression via angiogenesis inhibition could potentially augment the immunotherapeutic activity of immune checkpoint inhibitors,” the investigators wrote.

They reported results from the dose finding (1b) phase including 13 patients and initial phase 2 expansion cohorts with a total of 124 patients.

The maximum tolerated dose of lenvatinib in combination with pembrolizumab was established as 20 mg/day.

At 24 weeks of follow-up, the ORR for 30 patients with RCC was 63%; two additional patients had responses after week 24, for a total ORR at study cutoff in this cohort of 70%. The median duration of response for these patients was 20 months, and the median progression-free survival (PFS) was 19.8 months. At the time of data cutoff for this analysis, 9 of the 30 patients with RCC were still on treatment.

For 23 patients with endometrial cancer, the 24-week and overall ORR were 52%, with a median duration of response not reached, and a median PFS of 9.7 months. Seven patients were still on treatment at data cutoff.

For 21 patients with melanoma, the 24-week and overall ORR were 48%, median duration of response was 12.5 months, and median PFS was 5.5 months. Two of the patients were still on treatment at data cutoff.

For the 22 patients with squamous cell cancer of the head and neck, the 24-week ORR was 36%, with two patients having a response after week 24 for a total ORR at data cutoff of 46%. The median duration of response was 8.2 months and the median PFS was 4.7 months. Three patients remained on treatment at data cutoff.

For 21 patients with NSCLC, the 24-week and overall ORR were 33%, the median duration of response was 10.9 months, and median PFS was 5.9 months. Six of the patients were still receiving treatment at data cutoff.

For 20 patients with urothelial cancer, the 24-week and overall ORR were 25%, with a median duration of response not reached, and a median PFS of 5.4 months. Three patients were still receiving the combination at the time of data cutoff.

Treatment related adverse events (TRAEs) occurred in 133 of all 137 patients enrolled in the two study phases. The adverse events were similar across all cohorts, with any grade of events including fatigue in 58%, diarrhea in 52%, hypertension in 47%, hypothyroidism in 42%, and decreased appetite in 39%.

The most frequent grade 3 or 4 TRAEs were hypertension in 20%, fatigue in 12%, diarrhea in 9%, proteinuria in 8%, and increased lipase levels in 7%.

In all, 85% of patients had a TRAE leading to lenvatinib dose reduction and/or interruption, and 13% required lenvatinib discontinuation.

Events leading to pembrolizumab dose interruption occurred in 45% of patients, and pembrolizumab discontinuation in 15%.

The study was sponsored by Eisai with collaboration from Merck Sharp & Dohme. Dr. Taylor disclosed a consulting or advisory role for Bristol-Myers Squibb, Eisai, Array BioPharma, Loxo, Bayer, ArQule, Blueprint Medicines, Novartis, and Sanofi/Genzyme, and speakers bureau activities for BMS and Eisai.

SOURCE: Taylor MH et al. J Clin Oncol. 2020 Jan. 21 doi: 10.1200/JCO.19.01598.

 

A combination of the tyrosine kinase inhibitor lenvatinib (Lenvima) and the immune checkpoint inhibitor pembrolizumab (Keytruda) was safe and showed promising activity against advanced renal cell carcinoma and other solid tumors in a phase 1b/2 study.

Overall response rates (ORR) at 24 weeks ranged from 63% for patients with advanced renal cell carcinomas (RCC) to 25% for patients with urothelial cancers, reported Matthew H. Taylor, MD, of Knight Cancer Institute at Oregon Health & Science University in Portland, and colleagues.

The findings from this study sparked additional clinical trials for patients with gastric cancer, gastroesophageal cancer, and differentiated thyroid cancer, and set the stage for larger phase 3 trials in patients with advanced RCC, endometrial cancer, malignant melanoma, and non–small cell lung cancer (NSCLC).

“In the future, we also plan to study lenvatinib plus pembrolizumab in patients with RCC who have had disease progression after treatment with immune checkpoint inhibitors,” they wrote. The report was published in Journal of Clinical Oncology.

Lenvatinib is a multitargeted tyrosine kinase inhibitor (TKI) with action against vascular endothelial growth factor (VEGF) receptors 1-3, fibroblast growth factor (FGF) receptors 1-4, platelet-derived growth factor receptors alpha and the RET and KIT kinases.

“Preclinical and clinical studies suggest that modulation of VEGF-mediated immune suppression via angiogenesis inhibition could potentially augment the immunotherapeutic activity of immune checkpoint inhibitors,” the investigators wrote.

They reported results from the dose finding (1b) phase including 13 patients and initial phase 2 expansion cohorts with a total of 124 patients.

The maximum tolerated dose of lenvatinib in combination with pembrolizumab was established as 20 mg/day.

At 24 weeks of follow-up, the ORR for 30 patients with RCC was 63%; two additional patients had responses after week 24, for a total ORR at study cutoff in this cohort of 70%. The median duration of response for these patients was 20 months, and the median progression-free survival (PFS) was 19.8 months. At the time of data cutoff for this analysis, 9 of the 30 patients with RCC were still on treatment.

For 23 patients with endometrial cancer, the 24-week and overall ORR were 52%, with a median duration of response not reached, and a median PFS of 9.7 months. Seven patients were still on treatment at data cutoff.

For 21 patients with melanoma, the 24-week and overall ORR were 48%, median duration of response was 12.5 months, and median PFS was 5.5 months. Two of the patients were still on treatment at data cutoff.

For the 22 patients with squamous cell cancer of the head and neck, the 24-week ORR was 36%, with two patients having a response after week 24 for a total ORR at data cutoff of 46%. The median duration of response was 8.2 months and the median PFS was 4.7 months. Three patients remained on treatment at data cutoff.

For 21 patients with NSCLC, the 24-week and overall ORR were 33%, the median duration of response was 10.9 months, and median PFS was 5.9 months. Six of the patients were still receiving treatment at data cutoff.

For 20 patients with urothelial cancer, the 24-week and overall ORR were 25%, with a median duration of response not reached, and a median PFS of 5.4 months. Three patients were still receiving the combination at the time of data cutoff.

Treatment related adverse events (TRAEs) occurred in 133 of all 137 patients enrolled in the two study phases. The adverse events were similar across all cohorts, with any grade of events including fatigue in 58%, diarrhea in 52%, hypertension in 47%, hypothyroidism in 42%, and decreased appetite in 39%.

The most frequent grade 3 or 4 TRAEs were hypertension in 20%, fatigue in 12%, diarrhea in 9%, proteinuria in 8%, and increased lipase levels in 7%.

In all, 85% of patients had a TRAE leading to lenvatinib dose reduction and/or interruption, and 13% required lenvatinib discontinuation.

Events leading to pembrolizumab dose interruption occurred in 45% of patients, and pembrolizumab discontinuation in 15%.

The study was sponsored by Eisai with collaboration from Merck Sharp & Dohme. Dr. Taylor disclosed a consulting or advisory role for Bristol-Myers Squibb, Eisai, Array BioPharma, Loxo, Bayer, ArQule, Blueprint Medicines, Novartis, and Sanofi/Genzyme, and speakers bureau activities for BMS and Eisai.

SOURCE: Taylor MH et al. J Clin Oncol. 2020 Jan. 21 doi: 10.1200/JCO.19.01598.

Publications
Publications
Topics
Article Type
Click for Credit Status
Ready
Sections
Article Source

FROM THE JOURNAL OF CLINICAL ONCOLOGY

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.

ASCO issues guidelines on genetic testing in epithelial ovarian cancer

Article Type
Changed
Thu, 01/30/2020 - 17:22

In new guidelines, the American Society of Clinical Oncology recommends offering germline genetic testing for BRCA1, BRCA2, and other ovarian cancer susceptibility genes to all women diagnosed with epithelial ovarian cancer, regardless of their clinical features or family history.

Testing should be offered at diagnosis or as soon as possible after that, Panagiotis A. Konstantinopoulos, MD, PhD, of the Dana-Farber Cancer Institute in Boston, and colleagues wrote in the Journal of Clinical Oncology.

For patients who do not carry a germline pathogenic or likely pathogenic BRCA1/2 variant, the guidelines recommend offering somatic tumor testing for BRCA1/2 pathogenic or likely pathogenic variants. This testing can be offered at the time of disease recurrence after up-front therapy.

The guidelines also recommend somatic tumor testing for mismatch repair deficiency in patients diagnosed with clear cell, endometrioid, or mucinous ovarian cancer. This testing may be offered to patients with other histologic types of epithelial ovarian cancer as well.

Genetic testing, as well as genetic risk evaluation and counseling, should be offered to first- or second-degree blood relatives of a patient with ovarian cancer and a known germline pathogenic cancer susceptibility gene variant, according to the guidelines.

According to the guidelines, genetic evaluations should be conducted in cooperation with other health care providers who are “familiar with the diagnosis and management of hereditary cancer syndromes to determine the most appropriate testing strategy and discuss implications of the findings.”

Patients with identified germline or somatic pathogenic or likely pathogenic BRCA1/2 variants should receive treatments approved for them by the Food and Drug Administration, according to the guidelines. The authors note that patients with these variants have responded well to FDA-approved poly (ADP-ribose) polymerase inhibitors, including niraparib (Zejula), olaparib (Lynparza), and rucaparib (Rubraca).

The guidelines also state that mismatch repair deficiency qualifies for FDA-approved treatment, so patients with recurrent epithelial ovarian cancer and mismatch repair deficiency should receive FDA-approved treatments under their labeled indications.

The guidelines note that clinical decisions should not be based on a variant of uncertain significance. When a patient has such a variant, “clinical features and family history should inform clinical decision making,” according to the guidelines.

Dr. Konstantinopoulos and colleagues formulated the guidelines after reviewing data from 19 studies, including 6 meta-analyses; 11 randomized, controlled trials; and 2 observational studies.

The authors reported relationships with a range of pharmaceutical companies, including those that market drugs for epithelial ovarian cancer.

SOURCE: Konstantinopoulos PA et al. J Clin Oncol. 2020 Jan 27. doi: 10.1200/JCO.19.02960.

Publications
Topics
Sections

In new guidelines, the American Society of Clinical Oncology recommends offering germline genetic testing for BRCA1, BRCA2, and other ovarian cancer susceptibility genes to all women diagnosed with epithelial ovarian cancer, regardless of their clinical features or family history.

Testing should be offered at diagnosis or as soon as possible after that, Panagiotis A. Konstantinopoulos, MD, PhD, of the Dana-Farber Cancer Institute in Boston, and colleagues wrote in the Journal of Clinical Oncology.

For patients who do not carry a germline pathogenic or likely pathogenic BRCA1/2 variant, the guidelines recommend offering somatic tumor testing for BRCA1/2 pathogenic or likely pathogenic variants. This testing can be offered at the time of disease recurrence after up-front therapy.

The guidelines also recommend somatic tumor testing for mismatch repair deficiency in patients diagnosed with clear cell, endometrioid, or mucinous ovarian cancer. This testing may be offered to patients with other histologic types of epithelial ovarian cancer as well.

Genetic testing, as well as genetic risk evaluation and counseling, should be offered to first- or second-degree blood relatives of a patient with ovarian cancer and a known germline pathogenic cancer susceptibility gene variant, according to the guidelines.

According to the guidelines, genetic evaluations should be conducted in cooperation with other health care providers who are “familiar with the diagnosis and management of hereditary cancer syndromes to determine the most appropriate testing strategy and discuss implications of the findings.”

Patients with identified germline or somatic pathogenic or likely pathogenic BRCA1/2 variants should receive treatments approved for them by the Food and Drug Administration, according to the guidelines. The authors note that patients with these variants have responded well to FDA-approved poly (ADP-ribose) polymerase inhibitors, including niraparib (Zejula), olaparib (Lynparza), and rucaparib (Rubraca).

The guidelines also state that mismatch repair deficiency qualifies for FDA-approved treatment, so patients with recurrent epithelial ovarian cancer and mismatch repair deficiency should receive FDA-approved treatments under their labeled indications.

The guidelines note that clinical decisions should not be based on a variant of uncertain significance. When a patient has such a variant, “clinical features and family history should inform clinical decision making,” according to the guidelines.

Dr. Konstantinopoulos and colleagues formulated the guidelines after reviewing data from 19 studies, including 6 meta-analyses; 11 randomized, controlled trials; and 2 observational studies.

The authors reported relationships with a range of pharmaceutical companies, including those that market drugs for epithelial ovarian cancer.

SOURCE: Konstantinopoulos PA et al. J Clin Oncol. 2020 Jan 27. doi: 10.1200/JCO.19.02960.

In new guidelines, the American Society of Clinical Oncology recommends offering germline genetic testing for BRCA1, BRCA2, and other ovarian cancer susceptibility genes to all women diagnosed with epithelial ovarian cancer, regardless of their clinical features or family history.

Testing should be offered at diagnosis or as soon as possible after that, Panagiotis A. Konstantinopoulos, MD, PhD, of the Dana-Farber Cancer Institute in Boston, and colleagues wrote in the Journal of Clinical Oncology.

For patients who do not carry a germline pathogenic or likely pathogenic BRCA1/2 variant, the guidelines recommend offering somatic tumor testing for BRCA1/2 pathogenic or likely pathogenic variants. This testing can be offered at the time of disease recurrence after up-front therapy.

The guidelines also recommend somatic tumor testing for mismatch repair deficiency in patients diagnosed with clear cell, endometrioid, or mucinous ovarian cancer. This testing may be offered to patients with other histologic types of epithelial ovarian cancer as well.

Genetic testing, as well as genetic risk evaluation and counseling, should be offered to first- or second-degree blood relatives of a patient with ovarian cancer and a known germline pathogenic cancer susceptibility gene variant, according to the guidelines.

According to the guidelines, genetic evaluations should be conducted in cooperation with other health care providers who are “familiar with the diagnosis and management of hereditary cancer syndromes to determine the most appropriate testing strategy and discuss implications of the findings.”

Patients with identified germline or somatic pathogenic or likely pathogenic BRCA1/2 variants should receive treatments approved for them by the Food and Drug Administration, according to the guidelines. The authors note that patients with these variants have responded well to FDA-approved poly (ADP-ribose) polymerase inhibitors, including niraparib (Zejula), olaparib (Lynparza), and rucaparib (Rubraca).

The guidelines also state that mismatch repair deficiency qualifies for FDA-approved treatment, so patients with recurrent epithelial ovarian cancer and mismatch repair deficiency should receive FDA-approved treatments under their labeled indications.

The guidelines note that clinical decisions should not be based on a variant of uncertain significance. When a patient has such a variant, “clinical features and family history should inform clinical decision making,” according to the guidelines.

Dr. Konstantinopoulos and colleagues formulated the guidelines after reviewing data from 19 studies, including 6 meta-analyses; 11 randomized, controlled trials; and 2 observational studies.

The authors reported relationships with a range of pharmaceutical companies, including those that market drugs for epithelial ovarian cancer.

SOURCE: Konstantinopoulos PA et al. J Clin Oncol. 2020 Jan 27. doi: 10.1200/JCO.19.02960.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM THE JOURNAL OF CLINICAL ONCOLOGY

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.

Decreased incidence, survival in low-grade serous ovarian cancer illustrate ‘diagnostic shift’

Article Type
Changed
Thu, 01/16/2020 - 14:04

Relative to serous borderline ovarian tumors, the proportion of low-grade serous ovarian cancers has decreased in recent years, paralleled by a trend toward decreased survival, results of a large, population-based study suggest.

The number of low-grade serous ovarian cancer (LGSOC) cases declined by almost 26% in the retrospective analysis of registry data from 1998 to 2000, with a relative decrease of survival by nearly 29% over that time period.

Those decreases in LGSOC incidence and survival may be explained by a “diagnosis shift” toward versus serous borderline ovarian tumors (SBOTs) over time, though it could also be due to earlier detection of SBOT, which may be a precursor lesion of LGSOC, said authors of the study, led by oncology researcher Koji Matsuo, MD, PhD, of the University of Southern California, Los Angeles.

Taken together, the findings support the need to distinguish between LGSOC and SBOT, given their “distinctly different” oncologic outcomes and treatment approaches, according to Dr. Matsuo and colleagues.

“Making a proper diagnosis for LGSOC versus SBOT is paramount, as it impacts surgical and postoperative management,” the authors wrote in their report on the study, which appears in Gynecologic Oncology.

The retrospective, population-based, observational study, based on data from the Surveillance, Epidemiology, and End Results program of the National Cancer Institute, included women with LGSOC or SBOT diagnosed between 1988 and 2000 in the United States. A total of 4,712 cases were analyzed.

The number of LGSOC from 23.2% in 1988 down to 17.2% by 2000, or a relative decrease of 25.9%, Dr. Matsuo and coauthors reported. The decrease was even more pronounced in stage I disease, with a relative decrease of 37.6% versus 21.1% for stage II-IV.

As compared with women with SBOT, women with LGSOC were more likely to be older, more likely to have stage II-IV disease at diagnosis, more likely to have undergone hysterectomy, and less likely to be residents of the western United States, results of a multivariable analysis showed.

There was a downward trend in 15-year overall survival among women with LGSOC, from 53.3% to 38.0% by 2000, representing a relative decrease of 28.7%, according to the investigators. By contrast, overall survival was unchanged in the SBOT cohort, at 76.7% in 1988 and 78.7% by 2000, for a relative increase of about 2.5%.

Gradual changes in diagnostic classification over time may explain these trends, according to the investigators.

That shift would have started with the recognition of borderline ovarian tumors as a separate entity by the World Health Organization until 1971, leading to an increasing number of SBOTs over time as diagnostic practice patterns gradually changed. Later, it would have been fueled by the recognition of LGSOC as a new classification in the mid-1980s, which may have accelerated the shift by enhanced discrimination of SBOT from other serous tumors, the investigators wrote.

On the other hand, since SBOT may be a precursor lesion to LGSOC, the shift in diagnosis over time could be caused at least in part by increasing use of transvaginal ultrasonography, leading to more early detection of SBOT before they could progress to LGGOC. “More study is necessary to identify if SBOT can progress to LGSOC,” Dr. Matsuo and coauthors wrote.

The study was funded by the Ensign Endowment for Gynecologic Cancer Research. Dr. Matsuo reported disclosures related to Chugai, Springer, and VBL Therapeutics. His coauthors provided additional disclosures related to Kiyatec, Merck, Biopath, M-Trap, Quantgene, Tesaro, GlaxoSmithKline, Celgene, Johnson & Johnson, Biogin, Clovis Oncology, Novartis, Elsevier, and UpToDate.

SOURCE: Matsuo K et al. Gynecol Oncol. 2020 Jan 15. doi: 10.1016/j.ygyno.2019.08.030.

Publications
Topics
Sections

Relative to serous borderline ovarian tumors, the proportion of low-grade serous ovarian cancers has decreased in recent years, paralleled by a trend toward decreased survival, results of a large, population-based study suggest.

The number of low-grade serous ovarian cancer (LGSOC) cases declined by almost 26% in the retrospective analysis of registry data from 1998 to 2000, with a relative decrease of survival by nearly 29% over that time period.

Those decreases in LGSOC incidence and survival may be explained by a “diagnosis shift” toward versus serous borderline ovarian tumors (SBOTs) over time, though it could also be due to earlier detection of SBOT, which may be a precursor lesion of LGSOC, said authors of the study, led by oncology researcher Koji Matsuo, MD, PhD, of the University of Southern California, Los Angeles.

Taken together, the findings support the need to distinguish between LGSOC and SBOT, given their “distinctly different” oncologic outcomes and treatment approaches, according to Dr. Matsuo and colleagues.

“Making a proper diagnosis for LGSOC versus SBOT is paramount, as it impacts surgical and postoperative management,” the authors wrote in their report on the study, which appears in Gynecologic Oncology.

The retrospective, population-based, observational study, based on data from the Surveillance, Epidemiology, and End Results program of the National Cancer Institute, included women with LGSOC or SBOT diagnosed between 1988 and 2000 in the United States. A total of 4,712 cases were analyzed.

The number of LGSOC from 23.2% in 1988 down to 17.2% by 2000, or a relative decrease of 25.9%, Dr. Matsuo and coauthors reported. The decrease was even more pronounced in stage I disease, with a relative decrease of 37.6% versus 21.1% for stage II-IV.

As compared with women with SBOT, women with LGSOC were more likely to be older, more likely to have stage II-IV disease at diagnosis, more likely to have undergone hysterectomy, and less likely to be residents of the western United States, results of a multivariable analysis showed.

There was a downward trend in 15-year overall survival among women with LGSOC, from 53.3% to 38.0% by 2000, representing a relative decrease of 28.7%, according to the investigators. By contrast, overall survival was unchanged in the SBOT cohort, at 76.7% in 1988 and 78.7% by 2000, for a relative increase of about 2.5%.

Gradual changes in diagnostic classification over time may explain these trends, according to the investigators.

That shift would have started with the recognition of borderline ovarian tumors as a separate entity by the World Health Organization until 1971, leading to an increasing number of SBOTs over time as diagnostic practice patterns gradually changed. Later, it would have been fueled by the recognition of LGSOC as a new classification in the mid-1980s, which may have accelerated the shift by enhanced discrimination of SBOT from other serous tumors, the investigators wrote.

On the other hand, since SBOT may be a precursor lesion to LGSOC, the shift in diagnosis over time could be caused at least in part by increasing use of transvaginal ultrasonography, leading to more early detection of SBOT before they could progress to LGGOC. “More study is necessary to identify if SBOT can progress to LGSOC,” Dr. Matsuo and coauthors wrote.

The study was funded by the Ensign Endowment for Gynecologic Cancer Research. Dr. Matsuo reported disclosures related to Chugai, Springer, and VBL Therapeutics. His coauthors provided additional disclosures related to Kiyatec, Merck, Biopath, M-Trap, Quantgene, Tesaro, GlaxoSmithKline, Celgene, Johnson & Johnson, Biogin, Clovis Oncology, Novartis, Elsevier, and UpToDate.

SOURCE: Matsuo K et al. Gynecol Oncol. 2020 Jan 15. doi: 10.1016/j.ygyno.2019.08.030.

Relative to serous borderline ovarian tumors, the proportion of low-grade serous ovarian cancers has decreased in recent years, paralleled by a trend toward decreased survival, results of a large, population-based study suggest.

The number of low-grade serous ovarian cancer (LGSOC) cases declined by almost 26% in the retrospective analysis of registry data from 1998 to 2000, with a relative decrease of survival by nearly 29% over that time period.

Those decreases in LGSOC incidence and survival may be explained by a “diagnosis shift” toward versus serous borderline ovarian tumors (SBOTs) over time, though it could also be due to earlier detection of SBOT, which may be a precursor lesion of LGSOC, said authors of the study, led by oncology researcher Koji Matsuo, MD, PhD, of the University of Southern California, Los Angeles.

Taken together, the findings support the need to distinguish between LGSOC and SBOT, given their “distinctly different” oncologic outcomes and treatment approaches, according to Dr. Matsuo and colleagues.

“Making a proper diagnosis for LGSOC versus SBOT is paramount, as it impacts surgical and postoperative management,” the authors wrote in their report on the study, which appears in Gynecologic Oncology.

The retrospective, population-based, observational study, based on data from the Surveillance, Epidemiology, and End Results program of the National Cancer Institute, included women with LGSOC or SBOT diagnosed between 1988 and 2000 in the United States. A total of 4,712 cases were analyzed.

The number of LGSOC from 23.2% in 1988 down to 17.2% by 2000, or a relative decrease of 25.9%, Dr. Matsuo and coauthors reported. The decrease was even more pronounced in stage I disease, with a relative decrease of 37.6% versus 21.1% for stage II-IV.

As compared with women with SBOT, women with LGSOC were more likely to be older, more likely to have stage II-IV disease at diagnosis, more likely to have undergone hysterectomy, and less likely to be residents of the western United States, results of a multivariable analysis showed.

There was a downward trend in 15-year overall survival among women with LGSOC, from 53.3% to 38.0% by 2000, representing a relative decrease of 28.7%, according to the investigators. By contrast, overall survival was unchanged in the SBOT cohort, at 76.7% in 1988 and 78.7% by 2000, for a relative increase of about 2.5%.

Gradual changes in diagnostic classification over time may explain these trends, according to the investigators.

That shift would have started with the recognition of borderline ovarian tumors as a separate entity by the World Health Organization until 1971, leading to an increasing number of SBOTs over time as diagnostic practice patterns gradually changed. Later, it would have been fueled by the recognition of LGSOC as a new classification in the mid-1980s, which may have accelerated the shift by enhanced discrimination of SBOT from other serous tumors, the investigators wrote.

On the other hand, since SBOT may be a precursor lesion to LGSOC, the shift in diagnosis over time could be caused at least in part by increasing use of transvaginal ultrasonography, leading to more early detection of SBOT before they could progress to LGGOC. “More study is necessary to identify if SBOT can progress to LGSOC,” Dr. Matsuo and coauthors wrote.

The study was funded by the Ensign Endowment for Gynecologic Cancer Research. Dr. Matsuo reported disclosures related to Chugai, Springer, and VBL Therapeutics. His coauthors provided additional disclosures related to Kiyatec, Merck, Biopath, M-Trap, Quantgene, Tesaro, GlaxoSmithKline, Celgene, Johnson & Johnson, Biogin, Clovis Oncology, Novartis, Elsevier, and UpToDate.

SOURCE: Matsuo K et al. Gynecol Oncol. 2020 Jan 15. doi: 10.1016/j.ygyno.2019.08.030.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM GYNECOLOGIC ONCOLOGY

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.

Sharp declines for lung cancer, melanoma deaths fuel record drop in cancer mortality

Article Type
Changed
Fri, 01/17/2020 - 13:45

Declines in death rates for lung cancer and melanoma have gained momentum in recent years, fueling a record drop in cancer mortality, the American Cancer Society says.

Dr. Jacques P. Fontaine

Lung cancer death rates, which were falling by 3% in men and 2% in women annually in 2008 through 2013, dropped by 5% in men and nearly 4% per year in women annually from 2013 to 2017, according to the society’s 2020 statistical report.

Those accelerating reductions in death rates helped fuel the biggest-ever single year decline in overall cancer mortality, of 2.2%, from 2016 to 2017, their report shows.

According to the investigators, the decline in melanoma death rates escalated to 6.9% per year among 20- to 49-year-olds over 2013-2017, compared with a decline of just 2.9% per year during 2006-2010. Likewise, the melanoma death rate decline was 7.2% annually for the more recent time period, compared with just 1.3% annually in the earlier time period. The finding was even more remarkable for those 65 years of age and older, according to investigators, since the declines in melanoma death rates reached 6.2% annually, compared with a 0.9% annual increase in the years before immunotherapy.

Smoking cessation has been the main driver of progress in cutting lung cancer death rates, according to the report, while in melanoma, death rates have dropped after the introduction of immune checkpoint inhibitors and targeted therapies.

By contrast, reductions in death rates have slowed for colorectal cancers and female breast cancers, and have stabilized for prostate cancer, Ms. Siegel and coauthors stated, adding that racial and geographic disparities persist in preventable cancers, including those of the lung and cervix.

“Increased investment in both the equitable application of existing cancer control interventions and basic and clinical research to further advance treatment options would undoubtedly accelerate progress against cancer,” said the investigators. The report appears in CA: A Cancer Journal for Clinicians.

While the decline in lung cancer death rates is good news, the disease remains a major killer, responsible for more deaths than breast, colorectal, and ovarian cancer combined, said Jacques P. Fontaine, MD, a thoracic surgeon at Moffitt Cancer Center in Tampa, Fla.

“Five-year survival rates are still around the 18%-20% range, which is much lower than breast and prostate cancer,” Dr. Fontaine said in an interview. “Nonetheless, we’ve made a little dent in that, and we’re improving.”

Two other factors that have helped spur that improvement, according to Dr. Fontaine, are the reduced incidence of squamous cell carcinomas, which are linked to smoking, and the increased use of lung cancer screening with low-dose computed tomography.

Squamous cell carcinomas tend to be a central rather than peripheral, which makes the tumors harder to resect: “Surgery is sometimes not an option, and even to this day in 2020, the single most effective treatment for lung cancer remains surgical resection,” said Dr. Fontaine.

Likewise, centrally located tumors may preclude giving high-dose radiation and may result in more “collateral damage” to healthy tissue, he added.

Landmark studies show that low-dose CT scans reduce lung cancer deaths by 20% or more; however, screening can have false-positive results that lead to unnecessary biopsies and other harms, suggesting that the procedures should be done in centers of excellence that provide high-quality, responsible screening for early lung cancer, Dr. Fontaine said.

While the drop in melanoma death rates is encouraging and, not surprising in light of new cutting-edge therapies, an ongoing unmet treatment need still exists, according to Vishal Anil Patel, MD, director of cutaneous oncology at the George Washington Cancer Center in Washington.

“We still have a lot to learn, and a way to go, because we’ve really just made the first breakthrough,” Dr. Patel said in an interview.

Dr. Vishal Patel


Mortality data for melanoma can be challenging to interpret, according to Dr. Patel, given that more widespread screening may increase the number of documented melanoma cases with a lower risk of mortality.

Nevertheless, it’s not surprising that advanced melanoma death rates have declined precipitously, said Dr. Patel, since the diseases carries a high tumor mutational burden, which may explain the improved efficacy of immune checkpoint inhibitors.

“Without a doubt, the reason that people are living longer and doing better with this disease is because of these cutting-edge treatments that provide patients options that previously had no options at all, or a tailored option personalized to their tumor and focusing on what the patient really needs,” Dr. Patel said.

That said, response rates remain lower from other cancers, sparking interest in combining current immunotherapies with costimulatory molecules that may further improve survival rates, according to Dr. Patel.

In 2020, 606,000 cancer deaths are projected, according to the American Cancer Society statistical report. Of those deaths, nearly 136,000 are attributable to cancers of the lung and bronchus, while melanoma of the skin accounts for nearly 7,000 deaths.

The report notes that variation in cancer incidence reflects geographical differences in medical detection practices and the prevalence of risk factors, such as smoking, obesity, and other health behaviors. “For example, lung cancer incidence and mortality rates in Kentucky, where smoking prevalence was historically highest, are 3 to 4 times higher than those in Utah, where it was lowest. Even in 2018, 1 in 4 residents of Kentucky, Arkansas, and West Virginia were current smokers compared with 1 in 10 in Utah and California,” the investigators wrote.

Cancer mortality rates have fallen 29% since 1991, translating into 2.9 million fewer cancer deaths, the report says.

Dr. Siegel and coauthors are employed by the American Cancer Society, which receives grants from private and corporate foundations, and their salaries are solely funded through the American Cancer Society, according to the report.

SOURCE: Siegel RL et al. CA Cancer J Clin. 2020;70(1):7-30. doi: 10.3322/caac.21590.

Publications
Topics
Sections

Declines in death rates for lung cancer and melanoma have gained momentum in recent years, fueling a record drop in cancer mortality, the American Cancer Society says.

Dr. Jacques P. Fontaine

Lung cancer death rates, which were falling by 3% in men and 2% in women annually in 2008 through 2013, dropped by 5% in men and nearly 4% per year in women annually from 2013 to 2017, according to the society’s 2020 statistical report.

Those accelerating reductions in death rates helped fuel the biggest-ever single year decline in overall cancer mortality, of 2.2%, from 2016 to 2017, their report shows.

According to the investigators, the decline in melanoma death rates escalated to 6.9% per year among 20- to 49-year-olds over 2013-2017, compared with a decline of just 2.9% per year during 2006-2010. Likewise, the melanoma death rate decline was 7.2% annually for the more recent time period, compared with just 1.3% annually in the earlier time period. The finding was even more remarkable for those 65 years of age and older, according to investigators, since the declines in melanoma death rates reached 6.2% annually, compared with a 0.9% annual increase in the years before immunotherapy.

Smoking cessation has been the main driver of progress in cutting lung cancer death rates, according to the report, while in melanoma, death rates have dropped after the introduction of immune checkpoint inhibitors and targeted therapies.

By contrast, reductions in death rates have slowed for colorectal cancers and female breast cancers, and have stabilized for prostate cancer, Ms. Siegel and coauthors stated, adding that racial and geographic disparities persist in preventable cancers, including those of the lung and cervix.

“Increased investment in both the equitable application of existing cancer control interventions and basic and clinical research to further advance treatment options would undoubtedly accelerate progress against cancer,” said the investigators. The report appears in CA: A Cancer Journal for Clinicians.

While the decline in lung cancer death rates is good news, the disease remains a major killer, responsible for more deaths than breast, colorectal, and ovarian cancer combined, said Jacques P. Fontaine, MD, a thoracic surgeon at Moffitt Cancer Center in Tampa, Fla.

“Five-year survival rates are still around the 18%-20% range, which is much lower than breast and prostate cancer,” Dr. Fontaine said in an interview. “Nonetheless, we’ve made a little dent in that, and we’re improving.”

Two other factors that have helped spur that improvement, according to Dr. Fontaine, are the reduced incidence of squamous cell carcinomas, which are linked to smoking, and the increased use of lung cancer screening with low-dose computed tomography.

Squamous cell carcinomas tend to be a central rather than peripheral, which makes the tumors harder to resect: “Surgery is sometimes not an option, and even to this day in 2020, the single most effective treatment for lung cancer remains surgical resection,” said Dr. Fontaine.

Likewise, centrally located tumors may preclude giving high-dose radiation and may result in more “collateral damage” to healthy tissue, he added.

Landmark studies show that low-dose CT scans reduce lung cancer deaths by 20% or more; however, screening can have false-positive results that lead to unnecessary biopsies and other harms, suggesting that the procedures should be done in centers of excellence that provide high-quality, responsible screening for early lung cancer, Dr. Fontaine said.

While the drop in melanoma death rates is encouraging and, not surprising in light of new cutting-edge therapies, an ongoing unmet treatment need still exists, according to Vishal Anil Patel, MD, director of cutaneous oncology at the George Washington Cancer Center in Washington.

“We still have a lot to learn, and a way to go, because we’ve really just made the first breakthrough,” Dr. Patel said in an interview.

Dr. Vishal Patel


Mortality data for melanoma can be challenging to interpret, according to Dr. Patel, given that more widespread screening may increase the number of documented melanoma cases with a lower risk of mortality.

Nevertheless, it’s not surprising that advanced melanoma death rates have declined precipitously, said Dr. Patel, since the diseases carries a high tumor mutational burden, which may explain the improved efficacy of immune checkpoint inhibitors.

“Without a doubt, the reason that people are living longer and doing better with this disease is because of these cutting-edge treatments that provide patients options that previously had no options at all, or a tailored option personalized to their tumor and focusing on what the patient really needs,” Dr. Patel said.

That said, response rates remain lower from other cancers, sparking interest in combining current immunotherapies with costimulatory molecules that may further improve survival rates, according to Dr. Patel.

In 2020, 606,000 cancer deaths are projected, according to the American Cancer Society statistical report. Of those deaths, nearly 136,000 are attributable to cancers of the lung and bronchus, while melanoma of the skin accounts for nearly 7,000 deaths.

The report notes that variation in cancer incidence reflects geographical differences in medical detection practices and the prevalence of risk factors, such as smoking, obesity, and other health behaviors. “For example, lung cancer incidence and mortality rates in Kentucky, where smoking prevalence was historically highest, are 3 to 4 times higher than those in Utah, where it was lowest. Even in 2018, 1 in 4 residents of Kentucky, Arkansas, and West Virginia were current smokers compared with 1 in 10 in Utah and California,” the investigators wrote.

Cancer mortality rates have fallen 29% since 1991, translating into 2.9 million fewer cancer deaths, the report says.

Dr. Siegel and coauthors are employed by the American Cancer Society, which receives grants from private and corporate foundations, and their salaries are solely funded through the American Cancer Society, according to the report.

SOURCE: Siegel RL et al. CA Cancer J Clin. 2020;70(1):7-30. doi: 10.3322/caac.21590.

Declines in death rates for lung cancer and melanoma have gained momentum in recent years, fueling a record drop in cancer mortality, the American Cancer Society says.

Dr. Jacques P. Fontaine

Lung cancer death rates, which were falling by 3% in men and 2% in women annually in 2008 through 2013, dropped by 5% in men and nearly 4% per year in women annually from 2013 to 2017, according to the society’s 2020 statistical report.

Those accelerating reductions in death rates helped fuel the biggest-ever single year decline in overall cancer mortality, of 2.2%, from 2016 to 2017, their report shows.

According to the investigators, the decline in melanoma death rates escalated to 6.9% per year among 20- to 49-year-olds over 2013-2017, compared with a decline of just 2.9% per year during 2006-2010. Likewise, the melanoma death rate decline was 7.2% annually for the more recent time period, compared with just 1.3% annually in the earlier time period. The finding was even more remarkable for those 65 years of age and older, according to investigators, since the declines in melanoma death rates reached 6.2% annually, compared with a 0.9% annual increase in the years before immunotherapy.

Smoking cessation has been the main driver of progress in cutting lung cancer death rates, according to the report, while in melanoma, death rates have dropped after the introduction of immune checkpoint inhibitors and targeted therapies.

By contrast, reductions in death rates have slowed for colorectal cancers and female breast cancers, and have stabilized for prostate cancer, Ms. Siegel and coauthors stated, adding that racial and geographic disparities persist in preventable cancers, including those of the lung and cervix.

“Increased investment in both the equitable application of existing cancer control interventions and basic and clinical research to further advance treatment options would undoubtedly accelerate progress against cancer,” said the investigators. The report appears in CA: A Cancer Journal for Clinicians.

While the decline in lung cancer death rates is good news, the disease remains a major killer, responsible for more deaths than breast, colorectal, and ovarian cancer combined, said Jacques P. Fontaine, MD, a thoracic surgeon at Moffitt Cancer Center in Tampa, Fla.

“Five-year survival rates are still around the 18%-20% range, which is much lower than breast and prostate cancer,” Dr. Fontaine said in an interview. “Nonetheless, we’ve made a little dent in that, and we’re improving.”

Two other factors that have helped spur that improvement, according to Dr. Fontaine, are the reduced incidence of squamous cell carcinomas, which are linked to smoking, and the increased use of lung cancer screening with low-dose computed tomography.

Squamous cell carcinomas tend to be a central rather than peripheral, which makes the tumors harder to resect: “Surgery is sometimes not an option, and even to this day in 2020, the single most effective treatment for lung cancer remains surgical resection,” said Dr. Fontaine.

Likewise, centrally located tumors may preclude giving high-dose radiation and may result in more “collateral damage” to healthy tissue, he added.

Landmark studies show that low-dose CT scans reduce lung cancer deaths by 20% or more; however, screening can have false-positive results that lead to unnecessary biopsies and other harms, suggesting that the procedures should be done in centers of excellence that provide high-quality, responsible screening for early lung cancer, Dr. Fontaine said.

While the drop in melanoma death rates is encouraging and, not surprising in light of new cutting-edge therapies, an ongoing unmet treatment need still exists, according to Vishal Anil Patel, MD, director of cutaneous oncology at the George Washington Cancer Center in Washington.

“We still have a lot to learn, and a way to go, because we’ve really just made the first breakthrough,” Dr. Patel said in an interview.

Dr. Vishal Patel


Mortality data for melanoma can be challenging to interpret, according to Dr. Patel, given that more widespread screening may increase the number of documented melanoma cases with a lower risk of mortality.

Nevertheless, it’s not surprising that advanced melanoma death rates have declined precipitously, said Dr. Patel, since the diseases carries a high tumor mutational burden, which may explain the improved efficacy of immune checkpoint inhibitors.

“Without a doubt, the reason that people are living longer and doing better with this disease is because of these cutting-edge treatments that provide patients options that previously had no options at all, or a tailored option personalized to their tumor and focusing on what the patient really needs,” Dr. Patel said.

That said, response rates remain lower from other cancers, sparking interest in combining current immunotherapies with costimulatory molecules that may further improve survival rates, according to Dr. Patel.

In 2020, 606,000 cancer deaths are projected, according to the American Cancer Society statistical report. Of those deaths, nearly 136,000 are attributable to cancers of the lung and bronchus, while melanoma of the skin accounts for nearly 7,000 deaths.

The report notes that variation in cancer incidence reflects geographical differences in medical detection practices and the prevalence of risk factors, such as smoking, obesity, and other health behaviors. “For example, lung cancer incidence and mortality rates in Kentucky, where smoking prevalence was historically highest, are 3 to 4 times higher than those in Utah, where it was lowest. Even in 2018, 1 in 4 residents of Kentucky, Arkansas, and West Virginia were current smokers compared with 1 in 10 in Utah and California,” the investigators wrote.

Cancer mortality rates have fallen 29% since 1991, translating into 2.9 million fewer cancer deaths, the report says.

Dr. Siegel and coauthors are employed by the American Cancer Society, which receives grants from private and corporate foundations, and their salaries are solely funded through the American Cancer Society, according to the report.

SOURCE: Siegel RL et al. CA Cancer J Clin. 2020;70(1):7-30. doi: 10.3322/caac.21590.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM CA: A CANCER JOURNAL FOR CLINICIANS

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.

Should secondary cytoreduction be performed for platinum-sensitive recurrent ovarian cancer?

Article Type
Changed
Wed, 02/05/2020 - 09:57

Coleman RL, Spirtos NM, Enserro D, et al. Secondary surgical cytoreduction for recurrent ovarian cancer. N Engl J Med. 2019;381:1929-1939.

EXPERT COMMENTARY

Ovarian cancer represents the most lethal gynecologic cancer, with an estimated 14,000 deaths in 2019.1 While the incidence of this disease is low in comparison to uterine cancer, the advanced stage at diagnosis portends poor prognosis. While stage is an independent risk factor for death, it is also a risk for recurrence, with more than 80% of women developing recurrent disease.2-4 Secondary cytoreduction remains an option for patients in which disease recurs; up until now this management option was driven by retrospective data.5

Details of the study

Coleman and colleagues conducted the Gynecologic Oncology Group (GOG) 0213 trial—a phase 3, multicenter, randomized clinical trial that included 485 women with recurrent ovarian cancer. The surgical objective of the trial was to determine whether secondary cytoreduction in operable, platinum-sensitive (PS) patients improved overall survival (OS).

Patients were eligible to participate in the surgical portion of the trial if they had PS measurable disease and had the intention to achieve complete gross resection. Women with ascites, evidence of extraabdominal disease, and “diffuse carcinomatosis” were excluded. The primary and secondary end points were OS and progression-free survival (PFS), respectively.

Results. There were no statistical differences between the surgery and no surgery groups with regard to median OS (50.6 months vs 64.7 months, respectively; hazard ratio [HR], 1.29; 95% confidence interval [CI], 0.97–1.72) or median PFS (18.9 months vs 16.2 months; HR, 0.82; 95% CI, 0.66 to 1.01). When comparing patients in which complete gross resection was achieved (150 patients vs 245 who did not receive surgery), there was only a statistical difference in PFS in favor of the surgical group (22.4 months vs 16.2 months; HR, 0.62; 95% CI, 0.48–0.80).

Of note, 67% of the patients who received surgery (63% intention-to-treat) were debulked to complete gross resection. There were 33% more patients with extraabdominal disease (10% vs 7% of total patients in each group) and 15% more patients with upper abdominal disease (40% vs 33% of total patients in each group) included in the surgical group. Finally, the median time to chemotherapy was 40 days in the surgery group versus 7 days in the no surgery group.

Continue to: Study strengths and weaknesses...

 

 

Study strengths and weaknesses

The authors deserve to be commended for this well-designed and laborious trial, which is the first of its kind. The strength of the study is its randomized design producing level I data.

Study weaknesses include lack of reporting of BRCA status and the impact of receiving targeted therapies after the trial was over. It is well established that BRCA-mutated patients have an independent survival advantage, even when taking into account platinum sensitivity.6-8BRCA status of the study population is not specifically addressed in this paper. The authors noted in the first GOG 0213 trial publication, which assessed bevacizumab in the recurrent setting, that BRCA status has an impact on patient outcomes. Subsequently, they state that they do not report BRCA status because “…its independent effect on response to an anti-angiogenesis agent was unknown,” but it clearly would affect survival analysis if unbalanced between groups.9

Similarly, in the introduction to their study, Coleman and colleagues list availability of maintenance therapy, for instance poly ADP (adenosine diphosphate–ribose) polymerase (PARP) inhibitors, as rationale for conducting their trial. They subsequently cite this as a possible reason that the median overall survival was 3 times longer than expected. However, they provide no data on which patients received maintenance therapy, which again could have drastically affected survival outcomes.10 They do report in the supplementary information that, when stratifying those receiving bevacizumab adjuvantly during the trial, the median OS was comparable between the surgical and nonsurgical groups (58.5 months vs 61.7 months).

The authors discuss the presence of patient selection bias as a weakness in the study. Selection bias is evident in this trial (as in many surgical trials) because patients with a limited volume of disease were selected to participate over those with large-volume disease. It is reasonable to conclude that this study is likely selecting patients with less aggressive tumor biology, not only evident by low-volume disease at recurrence but also by the 20.4-month median platinum-free interval in the surgical group, which certainly affects the trial’s validity. Despite being considered PS, the disease biology in a patient with a platinum-free interval of 20.4 months is surely different from the disease biology in a patient with a 6.4-month platinum-free interval; therefore, it is difficult to generalize these data to all PS recurrent ovarian cancer patients. Similarly, other research has suggested strict selection criteria, which was not apparent in this study’s methodology.11 While the number of metastatic sites were relatively equal between the surgery and no surgery groups, there were more patients in the surgical group with extraabdominal disease, which the authors used as an exclusion criterion.

Lastly, the time to treatment commencement in each arm, which was 40 days for the surgical arm and 7 days in the nonsurgical arm, could represent a flaw in this trial. While we expect a difference in duration to account for recovery time, many centers start chemotherapy as soon as 21 days after surgery, which is almost half of the median interval in the surgical group in this trial. While the authors address this by stating that they completed a landmark analysis, no data or information about what time points they used for the analysis are provided. They simply report an interquartile range of 28 to 51 days. It is hard to know what effect this may have had on the outcome.

WHAT THIS EVIDENCE MEANS FOR PRACTICE

This is the first randomized clinical trial conducted to assess whether secondary surgical cytoreduction is beneficial in PS recurrent ovarian cancer patients. It provides compelling evidence to critically evaluate whether surgical cytoreduction is appropriate in a similar patient population. However, we would recommend using caution applying these data to patients who have different platinum-free intervals or low-volume disease limited to the pelvis.

The trial is not without flaws, as the authors point out in their discussion, but currently, it is the best evidence afforded to gynecologic oncologists. There are multiple trials currently ongoing, including DESTOP-III, which had similar PFS results as GOG 0213. If consensus is reached with these 2 trials, we believe that secondary cytoreduction will be utilized far less often in patients with recurrent ovarian cancer and a long platinum-free interval, thereby changing the current treatment paradigm for these patients.

MICHAEL D. TOBONI, MD, MPH, AND DAVID G. MUTCH, MD

References
  1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin. 2019;69:7-34. 
  2. Parmar MK, Ledermann JA, Colombo N, et al. Paclitaxel plus platinum-based chemotherapy versus conventional platinum-based chemotherapy in women with relapsed ovarian cancer: the ICON4/AGO-OVAR-2.2 trial. Lancet. 2003;361:2099-2106. 
  3. International Collaborative Ovarian Neoplasm Group. Paclitaxel plus carboplatin versus standard chemotherapy with either single-agent carboplatin or cyclophosphamide, doxorubicin, and cisplatin in women with ovarian cancer: the ICON3 randomised trial. Lancet. 2002;360:505-515. 
  4. Mullen MM, Kuroki LM, Thaker PH. Novel treatment options in platinum-sensitive recurrent ovarian cancer: a review. Gynecol Oncol. 2019;152:416-425. 
  5. National Comprehensive Cancer Network. NCCN clinical practice guidelines in oncology: ovarian cancer. November 26, 2019. https://www.nccn.org/professionals/physician_gls/pdf/ovarian.pdf. Accessed December 18, 2019.  
  6. Cass I, Baldwin RL, Varkey T, et al. Improved survival in women with BRCA-associated ovarian carcinoma. Cancer. 2003;97:2187-2195. 
  7. Gallagher DJ, Konner JA, Bell-McGuinn KM, et al. Survival in epithelial ovarian cancer: a multivariate analysis incorporating BRCA mutation status and platinum sensitivity. Ann Oncol. 2011;22:1127-1132. 
  8. Sun C, Li N, Ding D, et al. The role of BRCA status on the prognosis of patients with epithelial ovarian cancer: a systematic review of the literature with a meta-analysis. PLoS One. 2014;9:e95285.  
  9. Coleman RL, Brady MF, Herzog TJ, et al. Bevacizumab and paclitaxel-carboplatin chemotherapy and secondary cytoreduction in recurrent, platinum-sensitive ovarian cancer (NRG Oncology/Gynecologic Oncology Group study GOG-0213): a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2017;18:779-791. 
  10. Coleman RL, Spirtos NM, Enserro D, et al. Secondary surgical cytoreduction for recurrent ovarian cancer. N Engl J Med. 2019;381:1929-1939. 
  11. Chi DS, McCaughty K, Diaz JP, et al. Guidelines and selection criteria for secondary cytoreductive surgery in patients with recurrent, platinum-sensitive epithelial ovarian carcinoma. Cancer. 2006;106:1933-1939.
Article PDF
Author and Disclosure Information

Michael D. Toboni, MD, MPH, is Fellow, Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University in St. Louis, St. Louis, Missouri.

David G. Mutch, MD, is Ira C. and Judith Gall Professor of Obstetrics and Gynecology and Vice Chair of Gynecology in the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine in St. Louis and Alvin J. Siteman Cancer Center. He serves on the OBG Management Board of Editors.

The authors report no financial relationships relevant to this article.

Issue
OBG Management - 32(1)
Publications
Topics
Page Number
48-50
Sections
Author and Disclosure Information

Michael D. Toboni, MD, MPH, is Fellow, Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University in St. Louis, St. Louis, Missouri.

David G. Mutch, MD, is Ira C. and Judith Gall Professor of Obstetrics and Gynecology and Vice Chair of Gynecology in the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine in St. Louis and Alvin J. Siteman Cancer Center. He serves on the OBG Management Board of Editors.

The authors report no financial relationships relevant to this article.

Author and Disclosure Information

Michael D. Toboni, MD, MPH, is Fellow, Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University in St. Louis, St. Louis, Missouri.

David G. Mutch, MD, is Ira C. and Judith Gall Professor of Obstetrics and Gynecology and Vice Chair of Gynecology in the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine in St. Louis and Alvin J. Siteman Cancer Center. He serves on the OBG Management Board of Editors.

The authors report no financial relationships relevant to this article.

Article PDF
Article PDF

Coleman RL, Spirtos NM, Enserro D, et al. Secondary surgical cytoreduction for recurrent ovarian cancer. N Engl J Med. 2019;381:1929-1939.

EXPERT COMMENTARY

Ovarian cancer represents the most lethal gynecologic cancer, with an estimated 14,000 deaths in 2019.1 While the incidence of this disease is low in comparison to uterine cancer, the advanced stage at diagnosis portends poor prognosis. While stage is an independent risk factor for death, it is also a risk for recurrence, with more than 80% of women developing recurrent disease.2-4 Secondary cytoreduction remains an option for patients in which disease recurs; up until now this management option was driven by retrospective data.5

Details of the study

Coleman and colleagues conducted the Gynecologic Oncology Group (GOG) 0213 trial—a phase 3, multicenter, randomized clinical trial that included 485 women with recurrent ovarian cancer. The surgical objective of the trial was to determine whether secondary cytoreduction in operable, platinum-sensitive (PS) patients improved overall survival (OS).

Patients were eligible to participate in the surgical portion of the trial if they had PS measurable disease and had the intention to achieve complete gross resection. Women with ascites, evidence of extraabdominal disease, and “diffuse carcinomatosis” were excluded. The primary and secondary end points were OS and progression-free survival (PFS), respectively.

Results. There were no statistical differences between the surgery and no surgery groups with regard to median OS (50.6 months vs 64.7 months, respectively; hazard ratio [HR], 1.29; 95% confidence interval [CI], 0.97–1.72) or median PFS (18.9 months vs 16.2 months; HR, 0.82; 95% CI, 0.66 to 1.01). When comparing patients in which complete gross resection was achieved (150 patients vs 245 who did not receive surgery), there was only a statistical difference in PFS in favor of the surgical group (22.4 months vs 16.2 months; HR, 0.62; 95% CI, 0.48–0.80).

Of note, 67% of the patients who received surgery (63% intention-to-treat) were debulked to complete gross resection. There were 33% more patients with extraabdominal disease (10% vs 7% of total patients in each group) and 15% more patients with upper abdominal disease (40% vs 33% of total patients in each group) included in the surgical group. Finally, the median time to chemotherapy was 40 days in the surgery group versus 7 days in the no surgery group.

Continue to: Study strengths and weaknesses...

 

 

Study strengths and weaknesses

The authors deserve to be commended for this well-designed and laborious trial, which is the first of its kind. The strength of the study is its randomized design producing level I data.

Study weaknesses include lack of reporting of BRCA status and the impact of receiving targeted therapies after the trial was over. It is well established that BRCA-mutated patients have an independent survival advantage, even when taking into account platinum sensitivity.6-8BRCA status of the study population is not specifically addressed in this paper. The authors noted in the first GOG 0213 trial publication, which assessed bevacizumab in the recurrent setting, that BRCA status has an impact on patient outcomes. Subsequently, they state that they do not report BRCA status because “…its independent effect on response to an anti-angiogenesis agent was unknown,” but it clearly would affect survival analysis if unbalanced between groups.9

Similarly, in the introduction to their study, Coleman and colleagues list availability of maintenance therapy, for instance poly ADP (adenosine diphosphate–ribose) polymerase (PARP) inhibitors, as rationale for conducting their trial. They subsequently cite this as a possible reason that the median overall survival was 3 times longer than expected. However, they provide no data on which patients received maintenance therapy, which again could have drastically affected survival outcomes.10 They do report in the supplementary information that, when stratifying those receiving bevacizumab adjuvantly during the trial, the median OS was comparable between the surgical and nonsurgical groups (58.5 months vs 61.7 months).

The authors discuss the presence of patient selection bias as a weakness in the study. Selection bias is evident in this trial (as in many surgical trials) because patients with a limited volume of disease were selected to participate over those with large-volume disease. It is reasonable to conclude that this study is likely selecting patients with less aggressive tumor biology, not only evident by low-volume disease at recurrence but also by the 20.4-month median platinum-free interval in the surgical group, which certainly affects the trial’s validity. Despite being considered PS, the disease biology in a patient with a platinum-free interval of 20.4 months is surely different from the disease biology in a patient with a 6.4-month platinum-free interval; therefore, it is difficult to generalize these data to all PS recurrent ovarian cancer patients. Similarly, other research has suggested strict selection criteria, which was not apparent in this study’s methodology.11 While the number of metastatic sites were relatively equal between the surgery and no surgery groups, there were more patients in the surgical group with extraabdominal disease, which the authors used as an exclusion criterion.

Lastly, the time to treatment commencement in each arm, which was 40 days for the surgical arm and 7 days in the nonsurgical arm, could represent a flaw in this trial. While we expect a difference in duration to account for recovery time, many centers start chemotherapy as soon as 21 days after surgery, which is almost half of the median interval in the surgical group in this trial. While the authors address this by stating that they completed a landmark analysis, no data or information about what time points they used for the analysis are provided. They simply report an interquartile range of 28 to 51 days. It is hard to know what effect this may have had on the outcome.

WHAT THIS EVIDENCE MEANS FOR PRACTICE

This is the first randomized clinical trial conducted to assess whether secondary surgical cytoreduction is beneficial in PS recurrent ovarian cancer patients. It provides compelling evidence to critically evaluate whether surgical cytoreduction is appropriate in a similar patient population. However, we would recommend using caution applying these data to patients who have different platinum-free intervals or low-volume disease limited to the pelvis.

The trial is not without flaws, as the authors point out in their discussion, but currently, it is the best evidence afforded to gynecologic oncologists. There are multiple trials currently ongoing, including DESTOP-III, which had similar PFS results as GOG 0213. If consensus is reached with these 2 trials, we believe that secondary cytoreduction will be utilized far less often in patients with recurrent ovarian cancer and a long platinum-free interval, thereby changing the current treatment paradigm for these patients.

MICHAEL D. TOBONI, MD, MPH, AND DAVID G. MUTCH, MD

Coleman RL, Spirtos NM, Enserro D, et al. Secondary surgical cytoreduction for recurrent ovarian cancer. N Engl J Med. 2019;381:1929-1939.

EXPERT COMMENTARY

Ovarian cancer represents the most lethal gynecologic cancer, with an estimated 14,000 deaths in 2019.1 While the incidence of this disease is low in comparison to uterine cancer, the advanced stage at diagnosis portends poor prognosis. While stage is an independent risk factor for death, it is also a risk for recurrence, with more than 80% of women developing recurrent disease.2-4 Secondary cytoreduction remains an option for patients in which disease recurs; up until now this management option was driven by retrospective data.5

Details of the study

Coleman and colleagues conducted the Gynecologic Oncology Group (GOG) 0213 trial—a phase 3, multicenter, randomized clinical trial that included 485 women with recurrent ovarian cancer. The surgical objective of the trial was to determine whether secondary cytoreduction in operable, platinum-sensitive (PS) patients improved overall survival (OS).

Patients were eligible to participate in the surgical portion of the trial if they had PS measurable disease and had the intention to achieve complete gross resection. Women with ascites, evidence of extraabdominal disease, and “diffuse carcinomatosis” were excluded. The primary and secondary end points were OS and progression-free survival (PFS), respectively.

Results. There were no statistical differences between the surgery and no surgery groups with regard to median OS (50.6 months vs 64.7 months, respectively; hazard ratio [HR], 1.29; 95% confidence interval [CI], 0.97–1.72) or median PFS (18.9 months vs 16.2 months; HR, 0.82; 95% CI, 0.66 to 1.01). When comparing patients in which complete gross resection was achieved (150 patients vs 245 who did not receive surgery), there was only a statistical difference in PFS in favor of the surgical group (22.4 months vs 16.2 months; HR, 0.62; 95% CI, 0.48–0.80).

Of note, 67% of the patients who received surgery (63% intention-to-treat) were debulked to complete gross resection. There were 33% more patients with extraabdominal disease (10% vs 7% of total patients in each group) and 15% more patients with upper abdominal disease (40% vs 33% of total patients in each group) included in the surgical group. Finally, the median time to chemotherapy was 40 days in the surgery group versus 7 days in the no surgery group.

Continue to: Study strengths and weaknesses...

 

 

Study strengths and weaknesses

The authors deserve to be commended for this well-designed and laborious trial, which is the first of its kind. The strength of the study is its randomized design producing level I data.

Study weaknesses include lack of reporting of BRCA status and the impact of receiving targeted therapies after the trial was over. It is well established that BRCA-mutated patients have an independent survival advantage, even when taking into account platinum sensitivity.6-8BRCA status of the study population is not specifically addressed in this paper. The authors noted in the first GOG 0213 trial publication, which assessed bevacizumab in the recurrent setting, that BRCA status has an impact on patient outcomes. Subsequently, they state that they do not report BRCA status because “…its independent effect on response to an anti-angiogenesis agent was unknown,” but it clearly would affect survival analysis if unbalanced between groups.9

Similarly, in the introduction to their study, Coleman and colleagues list availability of maintenance therapy, for instance poly ADP (adenosine diphosphate–ribose) polymerase (PARP) inhibitors, as rationale for conducting their trial. They subsequently cite this as a possible reason that the median overall survival was 3 times longer than expected. However, they provide no data on which patients received maintenance therapy, which again could have drastically affected survival outcomes.10 They do report in the supplementary information that, when stratifying those receiving bevacizumab adjuvantly during the trial, the median OS was comparable between the surgical and nonsurgical groups (58.5 months vs 61.7 months).

The authors discuss the presence of patient selection bias as a weakness in the study. Selection bias is evident in this trial (as in many surgical trials) because patients with a limited volume of disease were selected to participate over those with large-volume disease. It is reasonable to conclude that this study is likely selecting patients with less aggressive tumor biology, not only evident by low-volume disease at recurrence but also by the 20.4-month median platinum-free interval in the surgical group, which certainly affects the trial’s validity. Despite being considered PS, the disease biology in a patient with a platinum-free interval of 20.4 months is surely different from the disease biology in a patient with a 6.4-month platinum-free interval; therefore, it is difficult to generalize these data to all PS recurrent ovarian cancer patients. Similarly, other research has suggested strict selection criteria, which was not apparent in this study’s methodology.11 While the number of metastatic sites were relatively equal between the surgery and no surgery groups, there were more patients in the surgical group with extraabdominal disease, which the authors used as an exclusion criterion.

Lastly, the time to treatment commencement in each arm, which was 40 days for the surgical arm and 7 days in the nonsurgical arm, could represent a flaw in this trial. While we expect a difference in duration to account for recovery time, many centers start chemotherapy as soon as 21 days after surgery, which is almost half of the median interval in the surgical group in this trial. While the authors address this by stating that they completed a landmark analysis, no data or information about what time points they used for the analysis are provided. They simply report an interquartile range of 28 to 51 days. It is hard to know what effect this may have had on the outcome.

WHAT THIS EVIDENCE MEANS FOR PRACTICE

This is the first randomized clinical trial conducted to assess whether secondary surgical cytoreduction is beneficial in PS recurrent ovarian cancer patients. It provides compelling evidence to critically evaluate whether surgical cytoreduction is appropriate in a similar patient population. However, we would recommend using caution applying these data to patients who have different platinum-free intervals or low-volume disease limited to the pelvis.

The trial is not without flaws, as the authors point out in their discussion, but currently, it is the best evidence afforded to gynecologic oncologists. There are multiple trials currently ongoing, including DESTOP-III, which had similar PFS results as GOG 0213. If consensus is reached with these 2 trials, we believe that secondary cytoreduction will be utilized far less often in patients with recurrent ovarian cancer and a long platinum-free interval, thereby changing the current treatment paradigm for these patients.

MICHAEL D. TOBONI, MD, MPH, AND DAVID G. MUTCH, MD

References
  1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin. 2019;69:7-34. 
  2. Parmar MK, Ledermann JA, Colombo N, et al. Paclitaxel plus platinum-based chemotherapy versus conventional platinum-based chemotherapy in women with relapsed ovarian cancer: the ICON4/AGO-OVAR-2.2 trial. Lancet. 2003;361:2099-2106. 
  3. International Collaborative Ovarian Neoplasm Group. Paclitaxel plus carboplatin versus standard chemotherapy with either single-agent carboplatin or cyclophosphamide, doxorubicin, and cisplatin in women with ovarian cancer: the ICON3 randomised trial. Lancet. 2002;360:505-515. 
  4. Mullen MM, Kuroki LM, Thaker PH. Novel treatment options in platinum-sensitive recurrent ovarian cancer: a review. Gynecol Oncol. 2019;152:416-425. 
  5. National Comprehensive Cancer Network. NCCN clinical practice guidelines in oncology: ovarian cancer. November 26, 2019. https://www.nccn.org/professionals/physician_gls/pdf/ovarian.pdf. Accessed December 18, 2019.  
  6. Cass I, Baldwin RL, Varkey T, et al. Improved survival in women with BRCA-associated ovarian carcinoma. Cancer. 2003;97:2187-2195. 
  7. Gallagher DJ, Konner JA, Bell-McGuinn KM, et al. Survival in epithelial ovarian cancer: a multivariate analysis incorporating BRCA mutation status and platinum sensitivity. Ann Oncol. 2011;22:1127-1132. 
  8. Sun C, Li N, Ding D, et al. The role of BRCA status on the prognosis of patients with epithelial ovarian cancer: a systematic review of the literature with a meta-analysis. PLoS One. 2014;9:e95285.  
  9. Coleman RL, Brady MF, Herzog TJ, et al. Bevacizumab and paclitaxel-carboplatin chemotherapy and secondary cytoreduction in recurrent, platinum-sensitive ovarian cancer (NRG Oncology/Gynecologic Oncology Group study GOG-0213): a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2017;18:779-791. 
  10. Coleman RL, Spirtos NM, Enserro D, et al. Secondary surgical cytoreduction for recurrent ovarian cancer. N Engl J Med. 2019;381:1929-1939. 
  11. Chi DS, McCaughty K, Diaz JP, et al. Guidelines and selection criteria for secondary cytoreductive surgery in patients with recurrent, platinum-sensitive epithelial ovarian carcinoma. Cancer. 2006;106:1933-1939.
References
  1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin. 2019;69:7-34. 
  2. Parmar MK, Ledermann JA, Colombo N, et al. Paclitaxel plus platinum-based chemotherapy versus conventional platinum-based chemotherapy in women with relapsed ovarian cancer: the ICON4/AGO-OVAR-2.2 trial. Lancet. 2003;361:2099-2106. 
  3. International Collaborative Ovarian Neoplasm Group. Paclitaxel plus carboplatin versus standard chemotherapy with either single-agent carboplatin or cyclophosphamide, doxorubicin, and cisplatin in women with ovarian cancer: the ICON3 randomised trial. Lancet. 2002;360:505-515. 
  4. Mullen MM, Kuroki LM, Thaker PH. Novel treatment options in platinum-sensitive recurrent ovarian cancer: a review. Gynecol Oncol. 2019;152:416-425. 
  5. National Comprehensive Cancer Network. NCCN clinical practice guidelines in oncology: ovarian cancer. November 26, 2019. https://www.nccn.org/professionals/physician_gls/pdf/ovarian.pdf. Accessed December 18, 2019.  
  6. Cass I, Baldwin RL, Varkey T, et al. Improved survival in women with BRCA-associated ovarian carcinoma. Cancer. 2003;97:2187-2195. 
  7. Gallagher DJ, Konner JA, Bell-McGuinn KM, et al. Survival in epithelial ovarian cancer: a multivariate analysis incorporating BRCA mutation status and platinum sensitivity. Ann Oncol. 2011;22:1127-1132. 
  8. Sun C, Li N, Ding D, et al. The role of BRCA status on the prognosis of patients with epithelial ovarian cancer: a systematic review of the literature with a meta-analysis. PLoS One. 2014;9:e95285.  
  9. Coleman RL, Brady MF, Herzog TJ, et al. Bevacizumab and paclitaxel-carboplatin chemotherapy and secondary cytoreduction in recurrent, platinum-sensitive ovarian cancer (NRG Oncology/Gynecologic Oncology Group study GOG-0213): a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2017;18:779-791. 
  10. Coleman RL, Spirtos NM, Enserro D, et al. Secondary surgical cytoreduction for recurrent ovarian cancer. N Engl J Med. 2019;381:1929-1939. 
  11. Chi DS, McCaughty K, Diaz JP, et al. Guidelines and selection criteria for secondary cytoreductive surgery in patients with recurrent, platinum-sensitive epithelial ovarian carcinoma. Cancer. 2006;106:1933-1939.
Issue
OBG Management - 32(1)
Issue
OBG Management - 32(1)
Page Number
48-50
Page Number
48-50
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.
Article PDF Media

Pelvic insufficiency fractures are common after chemoradiotherapy for cervical cancer

Article Type
Changed
Fri, 01/03/2020 - 12:35

– Radiation therapy for cervical cancer resulted in pelvic insufficiency fractures more frequently than previously thought, and many fractures were slow to heal, according to research presented at the annual meeting of the Radiological Society of North America.

“Pelvic insufficiency fractures had a prevalence of 38% on MRI follow-up” after chemoradiotherapy for locally advanced cervical cancer, said Alina Dragan, MD. This figure is more than double the previously reported prevalence of about 14%.

Dr. Dragan, a radiology resident at London North West Healthcare, National Health Service Trust, and coinvestigators also tracked the natural history of these fractures over time, to fill a knowledge gap about whether, and at what rate, these pelvic insufficiency fractures healed.

In the single-center retrospective study, the investigators found that just 14% of sacral fractures healed during the period of observation. For acetabular and pubic fractures, roughly one in three fractures had healed by the last MRI scan. About a third of all fractures remained stable across scans, while just over 10% of fractures were either fluctuant or worsened.

The study included 115 women with locally advanced cervical cancer who were treated with radical or adjuvant concurrent chemoradiotherapy over a 5-year period, and had MRI scans performed in-house; the follow-up protocol had patients receiving scans at 3, 12, and 24 months post treatment. From an initial pool of 197 patients, those who had previously had pelvic radiation or were receiving palliative treatment, as well as those with incomplete imaging follow-up and those with metal implants or prostheses that could affect radiation therapy delivery or imaging quality were excluded.

The chemoradiotherapy protocol involved five doses of weekly cisplatin at 400 mg/m2 of body surface area, as well as high–dose rate cervix brachytherapy. In practice, all but six participants received these treatments. Patients also received external beam radiotherapy with or without a simultaneous integrated boost to target affected lymph nodes, as clinically indicated.

The fractures were graded as mild, moderate, or severe by the interpreting radiologist according to the course of the fracture line and corresponding bone edema.

Patients were aged a median of 54 years, and 64 (56%) were postmenopausal. Most patients (n = 84; 73%) had never used tobacco. Participants’ median body mass index was 26 kg/m2.

Most patients (n = 73; 64%) were International Federation of Gynecology and Obstetrics stage 2b, and almost half (n = 55; 48%) had pelvic nodal involvement.

Patients were followed for a median of 12 months, with patients receiving a median of two MRIs curing that period. In all, 105 fractures were identified in 44 patients. A median of two fractures were identified among the group of patients who had pelvic insufficiency fractures.

The fractures were graded as mild, moderate, or severe by the interpreting radiologist according to the course of the fracture line and corresponding bone edema. In this schema, 41% of identified fractures were considered mild, while 32% were moderate and 12% were severe.

Although just over two-thirds of fractures (70%) were identified within 6 months of beginning surveillance, a quarter were not identified until 9-13 months post therapy, and 5% were found after more than 13 months.

Sacral fractures accounted for 72% of those identified, in keeping with previous findings, said Dr. Dragan. Acetabular and pubic fractures made up 16% and 10% of fractures, respectively. One fracture was seen at the ilium and one at the ischium.

Dr. Dragan and colleagues turned to multivariable analysis to look for risk factors for pelvic insufficiency fractures in this cohort of cervical cancer patients. Younger patients had a hazard ratio of 0.30 for fracture, compared with those over the age of 50 years (P less than .01). Similarly, being menopausal carried a hazard ratio of 2.25 for fracture. Higher radiation doses to the sacrum also boosted fracture risk (HR, 2.00; P = .03). Neither sacral volume and slope nor the receipt of simultaneous integrated boost were associated with increased fracture risk.

Dr. Dragan reported that she had no relevant conflicts of interest. She reported no outside sources of funding.
 

SOURCE: Dragan A et al. RSNA 2019, Presentation SSE25-03.

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

– Radiation therapy for cervical cancer resulted in pelvic insufficiency fractures more frequently than previously thought, and many fractures were slow to heal, according to research presented at the annual meeting of the Radiological Society of North America.

“Pelvic insufficiency fractures had a prevalence of 38% on MRI follow-up” after chemoradiotherapy for locally advanced cervical cancer, said Alina Dragan, MD. This figure is more than double the previously reported prevalence of about 14%.

Dr. Dragan, a radiology resident at London North West Healthcare, National Health Service Trust, and coinvestigators also tracked the natural history of these fractures over time, to fill a knowledge gap about whether, and at what rate, these pelvic insufficiency fractures healed.

In the single-center retrospective study, the investigators found that just 14% of sacral fractures healed during the period of observation. For acetabular and pubic fractures, roughly one in three fractures had healed by the last MRI scan. About a third of all fractures remained stable across scans, while just over 10% of fractures were either fluctuant or worsened.

The study included 115 women with locally advanced cervical cancer who were treated with radical or adjuvant concurrent chemoradiotherapy over a 5-year period, and had MRI scans performed in-house; the follow-up protocol had patients receiving scans at 3, 12, and 24 months post treatment. From an initial pool of 197 patients, those who had previously had pelvic radiation or were receiving palliative treatment, as well as those with incomplete imaging follow-up and those with metal implants or prostheses that could affect radiation therapy delivery or imaging quality were excluded.

The chemoradiotherapy protocol involved five doses of weekly cisplatin at 400 mg/m2 of body surface area, as well as high–dose rate cervix brachytherapy. In practice, all but six participants received these treatments. Patients also received external beam radiotherapy with or without a simultaneous integrated boost to target affected lymph nodes, as clinically indicated.

The fractures were graded as mild, moderate, or severe by the interpreting radiologist according to the course of the fracture line and corresponding bone edema.

Patients were aged a median of 54 years, and 64 (56%) were postmenopausal. Most patients (n = 84; 73%) had never used tobacco. Participants’ median body mass index was 26 kg/m2.

Most patients (n = 73; 64%) were International Federation of Gynecology and Obstetrics stage 2b, and almost half (n = 55; 48%) had pelvic nodal involvement.

Patients were followed for a median of 12 months, with patients receiving a median of two MRIs curing that period. In all, 105 fractures were identified in 44 patients. A median of two fractures were identified among the group of patients who had pelvic insufficiency fractures.

The fractures were graded as mild, moderate, or severe by the interpreting radiologist according to the course of the fracture line and corresponding bone edema. In this schema, 41% of identified fractures were considered mild, while 32% were moderate and 12% were severe.

Although just over two-thirds of fractures (70%) were identified within 6 months of beginning surveillance, a quarter were not identified until 9-13 months post therapy, and 5% were found after more than 13 months.

Sacral fractures accounted for 72% of those identified, in keeping with previous findings, said Dr. Dragan. Acetabular and pubic fractures made up 16% and 10% of fractures, respectively. One fracture was seen at the ilium and one at the ischium.

Dr. Dragan and colleagues turned to multivariable analysis to look for risk factors for pelvic insufficiency fractures in this cohort of cervical cancer patients. Younger patients had a hazard ratio of 0.30 for fracture, compared with those over the age of 50 years (P less than .01). Similarly, being menopausal carried a hazard ratio of 2.25 for fracture. Higher radiation doses to the sacrum also boosted fracture risk (HR, 2.00; P = .03). Neither sacral volume and slope nor the receipt of simultaneous integrated boost were associated with increased fracture risk.

Dr. Dragan reported that she had no relevant conflicts of interest. She reported no outside sources of funding.
 

SOURCE: Dragan A et al. RSNA 2019, Presentation SSE25-03.

– Radiation therapy for cervical cancer resulted in pelvic insufficiency fractures more frequently than previously thought, and many fractures were slow to heal, according to research presented at the annual meeting of the Radiological Society of North America.

“Pelvic insufficiency fractures had a prevalence of 38% on MRI follow-up” after chemoradiotherapy for locally advanced cervical cancer, said Alina Dragan, MD. This figure is more than double the previously reported prevalence of about 14%.

Dr. Dragan, a radiology resident at London North West Healthcare, National Health Service Trust, and coinvestigators also tracked the natural history of these fractures over time, to fill a knowledge gap about whether, and at what rate, these pelvic insufficiency fractures healed.

In the single-center retrospective study, the investigators found that just 14% of sacral fractures healed during the period of observation. For acetabular and pubic fractures, roughly one in three fractures had healed by the last MRI scan. About a third of all fractures remained stable across scans, while just over 10% of fractures were either fluctuant or worsened.

The study included 115 women with locally advanced cervical cancer who were treated with radical or adjuvant concurrent chemoradiotherapy over a 5-year period, and had MRI scans performed in-house; the follow-up protocol had patients receiving scans at 3, 12, and 24 months post treatment. From an initial pool of 197 patients, those who had previously had pelvic radiation or were receiving palliative treatment, as well as those with incomplete imaging follow-up and those with metal implants or prostheses that could affect radiation therapy delivery or imaging quality were excluded.

The chemoradiotherapy protocol involved five doses of weekly cisplatin at 400 mg/m2 of body surface area, as well as high–dose rate cervix brachytherapy. In practice, all but six participants received these treatments. Patients also received external beam radiotherapy with or without a simultaneous integrated boost to target affected lymph nodes, as clinically indicated.

The fractures were graded as mild, moderate, or severe by the interpreting radiologist according to the course of the fracture line and corresponding bone edema.

Patients were aged a median of 54 years, and 64 (56%) were postmenopausal. Most patients (n = 84; 73%) had never used tobacco. Participants’ median body mass index was 26 kg/m2.

Most patients (n = 73; 64%) were International Federation of Gynecology and Obstetrics stage 2b, and almost half (n = 55; 48%) had pelvic nodal involvement.

Patients were followed for a median of 12 months, with patients receiving a median of two MRIs curing that period. In all, 105 fractures were identified in 44 patients. A median of two fractures were identified among the group of patients who had pelvic insufficiency fractures.

The fractures were graded as mild, moderate, or severe by the interpreting radiologist according to the course of the fracture line and corresponding bone edema. In this schema, 41% of identified fractures were considered mild, while 32% were moderate and 12% were severe.

Although just over two-thirds of fractures (70%) were identified within 6 months of beginning surveillance, a quarter were not identified until 9-13 months post therapy, and 5% were found after more than 13 months.

Sacral fractures accounted for 72% of those identified, in keeping with previous findings, said Dr. Dragan. Acetabular and pubic fractures made up 16% and 10% of fractures, respectively. One fracture was seen at the ilium and one at the ischium.

Dr. Dragan and colleagues turned to multivariable analysis to look for risk factors for pelvic insufficiency fractures in this cohort of cervical cancer patients. Younger patients had a hazard ratio of 0.30 for fracture, compared with those over the age of 50 years (P less than .01). Similarly, being menopausal carried a hazard ratio of 2.25 for fracture. Higher radiation doses to the sacrum also boosted fracture risk (HR, 2.00; P = .03). Neither sacral volume and slope nor the receipt of simultaneous integrated boost were associated with increased fracture risk.

Dr. Dragan reported that she had no relevant conflicts of interest. She reported no outside sources of funding.
 

SOURCE: Dragan A et al. RSNA 2019, Presentation SSE25-03.

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM RSNA 2019

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.

Fertility-sparing surgery is safe for most with epithelial ovarian cancer

Article Type
Changed
Sun, 01/05/2020 - 09:30

Most young women with epithelial ovarian cancer can undergo surgery preserving the unaffected ovary and uterus – and thus their fertility – without compromising their survival, a cohort study of more than 9,000 women in Cancer suggests.

“Loss of reproductive capability and surgical menopause can negatively affect survivorship and quality of life among young women with ovarian cancer,” noted the investigators, who were led by Sarah M. Crafton, MD, division of gynecologic oncology, Allegheny Health Network in Pittsburgh. “ASCO has published guidelines to address the importance of implementing fertility preservation counseling as standard of care for all patients of reproductive age with cancer. However, the safety of such procedures should be thoroughly assessed in ongoing analyses.”

Dr. Crafton and colleagues used the Surveillance, Epidemiology, and End Results (SEER) program database and the National Cancer Database (NCDB) to retrospectively identify women 44 years old or younger with a primary epithelial ovarian cancer. The women were classified as having undergone surgery that spared fertility (unilateral salpingo-oophorectomy with uterine preservation) or surgery that did not (bilateral salpingo-oophorectomy with hysterectomy).

Study results, reported in Cancer, were based on 9,017 women – 3,932 from the SEER database and 5,085 from the NCDB – with epithelial ovarian cancer diagnosed between the ages of 15 and 44 years. Median follow-up was 6.5 years in SEER and 4.6 years in NCDB.

Overall, 26.1% of the SEER cohort and 24.8% of the NCDB cohort had undergone fertility-sparing surgery. In both cohorts, odds of this surgery were higher among younger women, those with a more recent ovarian cancer diagnosis, and those who did not receive adjuvant chemotherapy.

Among women with stage II-IV serous epithelial ovarian cancer in the SEER cohort, those who underwent fertility-sparing surgery had poorer overall survival (hazard ratio for death, 1.61; P = .0008). However, fertility-sparing surgery was not significantly associated with survival in other SEER subgroups defined by stage and grade or by stage and histology or in any NCDB subgroup defined by these parameters.

“In general, our findings regarding survival support the current National Comprehensive Cancer Network recommendation that fertility-sparing surgery can be considered as an alternative for traditional, comprehensive staging for those patients who desire fertility, for whom ovarian retention is technically feasible, and who have early-stage disease,” Dr. Crafton and coinvestigators wrote.

“Our observation of an increased risk of death associated with fertility-sparing surgery among women with advanced-stage, serous epithelial ovarian cancer in the SEER population supports the clinical recommendation that the decision to pursue fertility-sparing surgery should be individualized on the basis of patient/provider counseling and disease characteristics,” they concluded.

Dr. Crafton did not report any disclosures. The study was supported by the National Cancer Institute.

SOURCE: Crafton SM et al. Cancer. 2019 Nov 27. doi: 10.1002/cncr.32620.

Publications
Topics
Sections

Most young women with epithelial ovarian cancer can undergo surgery preserving the unaffected ovary and uterus – and thus their fertility – without compromising their survival, a cohort study of more than 9,000 women in Cancer suggests.

“Loss of reproductive capability and surgical menopause can negatively affect survivorship and quality of life among young women with ovarian cancer,” noted the investigators, who were led by Sarah M. Crafton, MD, division of gynecologic oncology, Allegheny Health Network in Pittsburgh. “ASCO has published guidelines to address the importance of implementing fertility preservation counseling as standard of care for all patients of reproductive age with cancer. However, the safety of such procedures should be thoroughly assessed in ongoing analyses.”

Dr. Crafton and colleagues used the Surveillance, Epidemiology, and End Results (SEER) program database and the National Cancer Database (NCDB) to retrospectively identify women 44 years old or younger with a primary epithelial ovarian cancer. The women were classified as having undergone surgery that spared fertility (unilateral salpingo-oophorectomy with uterine preservation) or surgery that did not (bilateral salpingo-oophorectomy with hysterectomy).

Study results, reported in Cancer, were based on 9,017 women – 3,932 from the SEER database and 5,085 from the NCDB – with epithelial ovarian cancer diagnosed between the ages of 15 and 44 years. Median follow-up was 6.5 years in SEER and 4.6 years in NCDB.

Overall, 26.1% of the SEER cohort and 24.8% of the NCDB cohort had undergone fertility-sparing surgery. In both cohorts, odds of this surgery were higher among younger women, those with a more recent ovarian cancer diagnosis, and those who did not receive adjuvant chemotherapy.

Among women with stage II-IV serous epithelial ovarian cancer in the SEER cohort, those who underwent fertility-sparing surgery had poorer overall survival (hazard ratio for death, 1.61; P = .0008). However, fertility-sparing surgery was not significantly associated with survival in other SEER subgroups defined by stage and grade or by stage and histology or in any NCDB subgroup defined by these parameters.

“In general, our findings regarding survival support the current National Comprehensive Cancer Network recommendation that fertility-sparing surgery can be considered as an alternative for traditional, comprehensive staging for those patients who desire fertility, for whom ovarian retention is technically feasible, and who have early-stage disease,” Dr. Crafton and coinvestigators wrote.

“Our observation of an increased risk of death associated with fertility-sparing surgery among women with advanced-stage, serous epithelial ovarian cancer in the SEER population supports the clinical recommendation that the decision to pursue fertility-sparing surgery should be individualized on the basis of patient/provider counseling and disease characteristics,” they concluded.

Dr. Crafton did not report any disclosures. The study was supported by the National Cancer Institute.

SOURCE: Crafton SM et al. Cancer. 2019 Nov 27. doi: 10.1002/cncr.32620.

Most young women with epithelial ovarian cancer can undergo surgery preserving the unaffected ovary and uterus – and thus their fertility – without compromising their survival, a cohort study of more than 9,000 women in Cancer suggests.

“Loss of reproductive capability and surgical menopause can negatively affect survivorship and quality of life among young women with ovarian cancer,” noted the investigators, who were led by Sarah M. Crafton, MD, division of gynecologic oncology, Allegheny Health Network in Pittsburgh. “ASCO has published guidelines to address the importance of implementing fertility preservation counseling as standard of care for all patients of reproductive age with cancer. However, the safety of such procedures should be thoroughly assessed in ongoing analyses.”

Dr. Crafton and colleagues used the Surveillance, Epidemiology, and End Results (SEER) program database and the National Cancer Database (NCDB) to retrospectively identify women 44 years old or younger with a primary epithelial ovarian cancer. The women were classified as having undergone surgery that spared fertility (unilateral salpingo-oophorectomy with uterine preservation) or surgery that did not (bilateral salpingo-oophorectomy with hysterectomy).

Study results, reported in Cancer, were based on 9,017 women – 3,932 from the SEER database and 5,085 from the NCDB – with epithelial ovarian cancer diagnosed between the ages of 15 and 44 years. Median follow-up was 6.5 years in SEER and 4.6 years in NCDB.

Overall, 26.1% of the SEER cohort and 24.8% of the NCDB cohort had undergone fertility-sparing surgery. In both cohorts, odds of this surgery were higher among younger women, those with a more recent ovarian cancer diagnosis, and those who did not receive adjuvant chemotherapy.

Among women with stage II-IV serous epithelial ovarian cancer in the SEER cohort, those who underwent fertility-sparing surgery had poorer overall survival (hazard ratio for death, 1.61; P = .0008). However, fertility-sparing surgery was not significantly associated with survival in other SEER subgroups defined by stage and grade or by stage and histology or in any NCDB subgroup defined by these parameters.

“In general, our findings regarding survival support the current National Comprehensive Cancer Network recommendation that fertility-sparing surgery can be considered as an alternative for traditional, comprehensive staging for those patients who desire fertility, for whom ovarian retention is technically feasible, and who have early-stage disease,” Dr. Crafton and coinvestigators wrote.

“Our observation of an increased risk of death associated with fertility-sparing surgery among women with advanced-stage, serous epithelial ovarian cancer in the SEER population supports the clinical recommendation that the decision to pursue fertility-sparing surgery should be individualized on the basis of patient/provider counseling and disease characteristics,” they concluded.

Dr. Crafton did not report any disclosures. The study was supported by the National Cancer Institute.

SOURCE: Crafton SM et al. Cancer. 2019 Nov 27. doi: 10.1002/cncr.32620.

Publications
Publications
Topics
Article Type
Click for Credit Status
Active
Sections
Article Source

FROM CANCER

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
CME ID
214359
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.