FDA approves idelalisib for CLL, SLL and FL

Article Type
Changed
Fri, 12/16/2022 - 12:26
Display Headline
FDA approves idelalisib for CLL, SLL and FL

The US Food and Drug Administration (FDA) has approved the PI3K delta inhibitor idelalisib (Zydelig) for the treatment of chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), and follicular lymphoma (FL).

 

The drug was granted traditional approval for use in combination with rituximab to treat patients with relapsed CLL who cannot receive rituximab alone.

 

Idelalisib has also received accelerated approval to treat patients with relapsed FL or SLL who have received at least 2 prior systemic therapies.

 

The FDA’s accelerated approval program allows for approval of a drug to treat a serious or life-threatening disease based on clinical data showing the drug has an effect on a surrogate endpoint that is reasonably likely to predict a clinical benefit to patients.

 

This program provides earlier patient access to a drug while the developer—in this case, Gilead Sciences—conducts trials confirming the drug’s benefit.

 

Idelalisib in CLL: Results of a phase 3 study

 

The approval of idelalisib in CLL is based on results of a phase 3 trial (Study 116), which was stopped early because idelalisib had a significant impact on progression-free survival.

 

The study included 220 CLL patients who could not receive chemotherapy. Half were randomized to receive idelalisib plus rituximab, and the other half were randomized to rituximab plus placebo.

 

Patients in the idelalisib arm had a much higher overall response rate than patients in the placebo arm—81% and 13%, respectively (P<0.001). But all responses were partial responses.

 

At 24 weeks, the rate of progression-free survival was 93% in the idelalisib arm and 46% in the placebo arm (P<0.001). The median progression-free survival was 5.5 months in the placebo arm and not reached in the idelalisib arm (P<0.001).

 

At 12 months, the overall survival rate was 92% in the idelalisib arm and 80% in the placebo arm (P=0.02).

 

Most adverse events, in either treatment group, were grade 2 or lower. The most common events in the idelalisib arm were pyrexia, fatigue, nausea, chills, and diarrhea. In the placebo arm, the most common events were infusion-related reactions, fatigue, cough, nausea, and dyspnea.

 

There were more serious adverse events in the idelalisib arm than in the placebo arm—40% and 35%, respectively. The most frequent serious events were pneumonia, pyrexia, and febrile neutropenia (in both treatment arms).

 

Idelalisib in FL and SLL: Results of a phase 2 study

 

Idelalisib’s accelerated approval in FL and SLL is supported by data from a single-arm, phase 2 trial (Study 101-09).

 

The drug was given as a single agent to patients who were refractory to rituximab and alkylating-agent-containing chemotherapy. Seventy-two patients had FL, and 26 had SLL.

 

The overall response rate was 54% in FL and 58% in SLL. Eight percent of FL responses were complete, and all responses in SLL patients were partial.

 

The median duration of response was 11.9 months in SLL patients (range, 0-14.7 months) but was not reached in FL patients (range, 0-14.8 months).

 

Improvements in patient survival or disease-related symptoms have not been established.

 

In all patients, the most common grade 3 or higher adverse events were neutropenia (27%), elevations in aminotransferase levels (13%), diarrhea (13%), and pneumonia (7%).

 

About idelalisib: Dosing, boxed warning and REMS

 

Idelalisib is an oral inhibitor of PI3K delta, a protein that plays a role in the activation, proliferation, and viability of B cells. PI3K delta signaling is active in many B-cell leukemias and lymphomas, and, by inhibiting the protein, idelalisib blocks several cellular signaling pathways that drive B-cell viability.

 

The drug is available as 150 mg and 100 mg tablets, administered orally twice-daily, but 150 mg is the recommended starting dose.

 

 

 

Idelalisib has a boxed warning on its label communicating the risks of fatal and serious toxicities, which include hepatic toxicity, severe diarrhea, colitis, pneumonitis, and intestinal perforation.

 

The drug is being approved with a risk evaluation and mitigation strategy (REMS) comprised of a communication plan to ensure healthcare providers are fully informed about these risks. For more information on this program, visit www.ZydeligREMS.com.

Publications
Topics

The US Food and Drug Administration (FDA) has approved the PI3K delta inhibitor idelalisib (Zydelig) for the treatment of chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), and follicular lymphoma (FL).

 

The drug was granted traditional approval for use in combination with rituximab to treat patients with relapsed CLL who cannot receive rituximab alone.

 

Idelalisib has also received accelerated approval to treat patients with relapsed FL or SLL who have received at least 2 prior systemic therapies.

 

The FDA’s accelerated approval program allows for approval of a drug to treat a serious or life-threatening disease based on clinical data showing the drug has an effect on a surrogate endpoint that is reasonably likely to predict a clinical benefit to patients.

 

This program provides earlier patient access to a drug while the developer—in this case, Gilead Sciences—conducts trials confirming the drug’s benefit.

 

Idelalisib in CLL: Results of a phase 3 study

 

The approval of idelalisib in CLL is based on results of a phase 3 trial (Study 116), which was stopped early because idelalisib had a significant impact on progression-free survival.

 

The study included 220 CLL patients who could not receive chemotherapy. Half were randomized to receive idelalisib plus rituximab, and the other half were randomized to rituximab plus placebo.

 

Patients in the idelalisib arm had a much higher overall response rate than patients in the placebo arm—81% and 13%, respectively (P<0.001). But all responses were partial responses.

 

At 24 weeks, the rate of progression-free survival was 93% in the idelalisib arm and 46% in the placebo arm (P<0.001). The median progression-free survival was 5.5 months in the placebo arm and not reached in the idelalisib arm (P<0.001).

 

At 12 months, the overall survival rate was 92% in the idelalisib arm and 80% in the placebo arm (P=0.02).

 

Most adverse events, in either treatment group, were grade 2 or lower. The most common events in the idelalisib arm were pyrexia, fatigue, nausea, chills, and diarrhea. In the placebo arm, the most common events were infusion-related reactions, fatigue, cough, nausea, and dyspnea.

 

There were more serious adverse events in the idelalisib arm than in the placebo arm—40% and 35%, respectively. The most frequent serious events were pneumonia, pyrexia, and febrile neutropenia (in both treatment arms).

 

Idelalisib in FL and SLL: Results of a phase 2 study

 

Idelalisib’s accelerated approval in FL and SLL is supported by data from a single-arm, phase 2 trial (Study 101-09).

 

The drug was given as a single agent to patients who were refractory to rituximab and alkylating-agent-containing chemotherapy. Seventy-two patients had FL, and 26 had SLL.

 

The overall response rate was 54% in FL and 58% in SLL. Eight percent of FL responses were complete, and all responses in SLL patients were partial.

 

The median duration of response was 11.9 months in SLL patients (range, 0-14.7 months) but was not reached in FL patients (range, 0-14.8 months).

 

Improvements in patient survival or disease-related symptoms have not been established.

 

In all patients, the most common grade 3 or higher adverse events were neutropenia (27%), elevations in aminotransferase levels (13%), diarrhea (13%), and pneumonia (7%).

 

About idelalisib: Dosing, boxed warning and REMS

 

Idelalisib is an oral inhibitor of PI3K delta, a protein that plays a role in the activation, proliferation, and viability of B cells. PI3K delta signaling is active in many B-cell leukemias and lymphomas, and, by inhibiting the protein, idelalisib blocks several cellular signaling pathways that drive B-cell viability.

 

The drug is available as 150 mg and 100 mg tablets, administered orally twice-daily, but 150 mg is the recommended starting dose.

 

 

 

Idelalisib has a boxed warning on its label communicating the risks of fatal and serious toxicities, which include hepatic toxicity, severe diarrhea, colitis, pneumonitis, and intestinal perforation.

 

The drug is being approved with a risk evaluation and mitigation strategy (REMS) comprised of a communication plan to ensure healthcare providers are fully informed about these risks. For more information on this program, visit www.ZydeligREMS.com.

The US Food and Drug Administration (FDA) has approved the PI3K delta inhibitor idelalisib (Zydelig) for the treatment of chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), and follicular lymphoma (FL).

 

The drug was granted traditional approval for use in combination with rituximab to treat patients with relapsed CLL who cannot receive rituximab alone.

 

Idelalisib has also received accelerated approval to treat patients with relapsed FL or SLL who have received at least 2 prior systemic therapies.

 

The FDA’s accelerated approval program allows for approval of a drug to treat a serious or life-threatening disease based on clinical data showing the drug has an effect on a surrogate endpoint that is reasonably likely to predict a clinical benefit to patients.

 

This program provides earlier patient access to a drug while the developer—in this case, Gilead Sciences—conducts trials confirming the drug’s benefit.

 

Idelalisib in CLL: Results of a phase 3 study

 

The approval of idelalisib in CLL is based on results of a phase 3 trial (Study 116), which was stopped early because idelalisib had a significant impact on progression-free survival.

 

The study included 220 CLL patients who could not receive chemotherapy. Half were randomized to receive idelalisib plus rituximab, and the other half were randomized to rituximab plus placebo.

 

Patients in the idelalisib arm had a much higher overall response rate than patients in the placebo arm—81% and 13%, respectively (P<0.001). But all responses were partial responses.

 

At 24 weeks, the rate of progression-free survival was 93% in the idelalisib arm and 46% in the placebo arm (P<0.001). The median progression-free survival was 5.5 months in the placebo arm and not reached in the idelalisib arm (P<0.001).

 

At 12 months, the overall survival rate was 92% in the idelalisib arm and 80% in the placebo arm (P=0.02).

 

Most adverse events, in either treatment group, were grade 2 or lower. The most common events in the idelalisib arm were pyrexia, fatigue, nausea, chills, and diarrhea. In the placebo arm, the most common events were infusion-related reactions, fatigue, cough, nausea, and dyspnea.

 

There were more serious adverse events in the idelalisib arm than in the placebo arm—40% and 35%, respectively. The most frequent serious events were pneumonia, pyrexia, and febrile neutropenia (in both treatment arms).

 

Idelalisib in FL and SLL: Results of a phase 2 study

 

Idelalisib’s accelerated approval in FL and SLL is supported by data from a single-arm, phase 2 trial (Study 101-09).

 

The drug was given as a single agent to patients who were refractory to rituximab and alkylating-agent-containing chemotherapy. Seventy-two patients had FL, and 26 had SLL.

 

The overall response rate was 54% in FL and 58% in SLL. Eight percent of FL responses were complete, and all responses in SLL patients were partial.

 

The median duration of response was 11.9 months in SLL patients (range, 0-14.7 months) but was not reached in FL patients (range, 0-14.8 months).

 

Improvements in patient survival or disease-related symptoms have not been established.

 

In all patients, the most common grade 3 or higher adverse events were neutropenia (27%), elevations in aminotransferase levels (13%), diarrhea (13%), and pneumonia (7%).

 

About idelalisib: Dosing, boxed warning and REMS

 

Idelalisib is an oral inhibitor of PI3K delta, a protein that plays a role in the activation, proliferation, and viability of B cells. PI3K delta signaling is active in many B-cell leukemias and lymphomas, and, by inhibiting the protein, idelalisib blocks several cellular signaling pathways that drive B-cell viability.

 

The drug is available as 150 mg and 100 mg tablets, administered orally twice-daily, but 150 mg is the recommended starting dose.

 

 

 

Idelalisib has a boxed warning on its label communicating the risks of fatal and serious toxicities, which include hepatic toxicity, severe diarrhea, colitis, pneumonitis, and intestinal perforation.

 

The drug is being approved with a risk evaluation and mitigation strategy (REMS) comprised of a communication plan to ensure healthcare providers are fully informed about these risks. For more information on this program, visit www.ZydeligREMS.com.

Publications
Publications
Topics
Article Type
Display Headline
FDA approves idelalisib for CLL, SLL and FL
Display Headline
FDA approves idelalisib for CLL, SLL and FL
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

FDA approves idelalisib for three leukemia and lymphoma indications

Article Type
Changed
Fri, 12/16/2022 - 12:26
Display Headline
FDA approves idelalisib for three leukemia and lymphoma indications

The oral kinase inhibitor idelalisib has been approved for treating patients with relapsed chronic lymphocytic leukemia, follicular lymphoma, and small lymphocytic lymphoma, with a boxed warning about fatal and serious toxicities associated with treatment, the Food and Drug Administration announced on July 23.

The three approved indications for idelalisib – administered at a recommended starting dose of 150 mg, twice a day – are for:

 

• Relapsed chronic lymphocytic leukemia (CLL), in combination with rituximab, in patients for whom rituximab alone would be considered appropriate therapy due to other comorbidities. This is a traditional approval based on progression-free survival (PFS) data.

• Relapsed follicular B-cell non-Hodgkin’s lymphoma (FL) in patients who have received at least two prior systemic therapies.

• Relapsed small lymphocytic lymphoma (SLL) in patients who have received at least two prior systemic therapies.

The FL and SLL indications, based on objective response rates in one study, are accelerated approvals, which provides patients with serious or life-threatening diseases earlier access to a promising drug based on an effect on a surrogate endpoint considered " reasonably likely to predict clinical benefit," according to the FDA. Full approval is contingent on the manufacturer conducting trials confirming clinical benefits; if they do not, the FDA can withdraw the approval.

Idelalisib is being marketed as Zydelig, by Gilead Sciences. There is also a Risk Evaluation and Mitigation Strategy (REMS) in place for the drug, which includes a communication plan to ensure that prescribers are fully informed about treatment-associated risks. The boxed warning lists the risks of fatal and/or serious hepatoxicity (affecting 14% of treated patients); fatal and/or serious and severe diarrhea or colitis (also affecting 14% of treated patients); as well as fatal and serious pneumonitis; and fatal and serious intestinal perforation.

Idelalisib is an oral inhibitor of phosphoinositide 3-kinase (PI3K) delta, "a protein that plays a role in the activation, proliferation, and viability of B cells," according to a Gilead statement announcing the approval, which added that PI3K delta signaling "is active in many B-cell leukemias and lymphomas, and by inhibiting the protein, Zydelig blocks several cellular signaling pathways that drive B-cell viability."

The FDA granted a full approval for the CLL indication, based on a phase III study of 220 patients, which was stopped early in October 2013 at the first-specified interim analysis. Median PFS was not reached among those randomized to on idelalisib plus rituximab, but was at least 10.7 months, and was 5.5 months among those randomized to placebo plus rituximab (N. Engl. J. Med. 2014;370:997-1007). "Results from a second interim analysis continued to show a statistically significant improvement for Zydelig and Rituxan over placebo and Rituxan," the FDA statement said.

The accelerated approvals for relapsed FL and relapsed SLL were based on a single-arm phase II study of 123 patients refractory to rituximab and chemotherapy including alkylating agents, treated with idelalisib. The objective response rates were 54% among those with relapsed FL, and 58% of those with SLL in the study (N. Engl. J. Med. 2014;370:1008-18).

Common adverse events associated with treatment included diarrhea, fever, fatigue, nausea, cough, pneumonia, abdominal pain, chills, and rash; and common lab abnormalities associated with treatment included neutropenia, hypertriglyceridemia, hyperglycemia, and liver enzyme elevations, according to the FDA.

In the FDA statement, Dr. Richard Pazdur, director of the Office of Hematology and Oncology Products in the FDA’s Center for Drug Evaluation and Research, pointed out that in less than a year, "We have seen considerable progress in the availability of treatments for chronic lymphocytic leukemia." The other treatments for CLL are obinutuzumab (Gazyva), approved in November 2013; ibrutinib (Imbruvica), approved in February 2014; and ofatumumab (Arzerra) approved in April 2014.

Prescribing information is available at the FDA website.

[email protected]

Publications
Topics
Legacy Keywords
oral kinase inhibitor, idelalisib, relapsed chronic lymphocytic leukemia, follicular lymphoma, small lymphocytic lymphoma, Food and Drug Administration,
Related Articles

The oral kinase inhibitor idelalisib has been approved for treating patients with relapsed chronic lymphocytic leukemia, follicular lymphoma, and small lymphocytic lymphoma, with a boxed warning about fatal and serious toxicities associated with treatment, the Food and Drug Administration announced on July 23.

The three approved indications for idelalisib – administered at a recommended starting dose of 150 mg, twice a day – are for:

 

• Relapsed chronic lymphocytic leukemia (CLL), in combination with rituximab, in patients for whom rituximab alone would be considered appropriate therapy due to other comorbidities. This is a traditional approval based on progression-free survival (PFS) data.

• Relapsed follicular B-cell non-Hodgkin’s lymphoma (FL) in patients who have received at least two prior systemic therapies.

• Relapsed small lymphocytic lymphoma (SLL) in patients who have received at least two prior systemic therapies.

The FL and SLL indications, based on objective response rates in one study, are accelerated approvals, which provides patients with serious or life-threatening diseases earlier access to a promising drug based on an effect on a surrogate endpoint considered " reasonably likely to predict clinical benefit," according to the FDA. Full approval is contingent on the manufacturer conducting trials confirming clinical benefits; if they do not, the FDA can withdraw the approval.

Idelalisib is being marketed as Zydelig, by Gilead Sciences. There is also a Risk Evaluation and Mitigation Strategy (REMS) in place for the drug, which includes a communication plan to ensure that prescribers are fully informed about treatment-associated risks. The boxed warning lists the risks of fatal and/or serious hepatoxicity (affecting 14% of treated patients); fatal and/or serious and severe diarrhea or colitis (also affecting 14% of treated patients); as well as fatal and serious pneumonitis; and fatal and serious intestinal perforation.

Idelalisib is an oral inhibitor of phosphoinositide 3-kinase (PI3K) delta, "a protein that plays a role in the activation, proliferation, and viability of B cells," according to a Gilead statement announcing the approval, which added that PI3K delta signaling "is active in many B-cell leukemias and lymphomas, and by inhibiting the protein, Zydelig blocks several cellular signaling pathways that drive B-cell viability."

The FDA granted a full approval for the CLL indication, based on a phase III study of 220 patients, which was stopped early in October 2013 at the first-specified interim analysis. Median PFS was not reached among those randomized to on idelalisib plus rituximab, but was at least 10.7 months, and was 5.5 months among those randomized to placebo plus rituximab (N. Engl. J. Med. 2014;370:997-1007). "Results from a second interim analysis continued to show a statistically significant improvement for Zydelig and Rituxan over placebo and Rituxan," the FDA statement said.

The accelerated approvals for relapsed FL and relapsed SLL were based on a single-arm phase II study of 123 patients refractory to rituximab and chemotherapy including alkylating agents, treated with idelalisib. The objective response rates were 54% among those with relapsed FL, and 58% of those with SLL in the study (N. Engl. J. Med. 2014;370:1008-18).

Common adverse events associated with treatment included diarrhea, fever, fatigue, nausea, cough, pneumonia, abdominal pain, chills, and rash; and common lab abnormalities associated with treatment included neutropenia, hypertriglyceridemia, hyperglycemia, and liver enzyme elevations, according to the FDA.

In the FDA statement, Dr. Richard Pazdur, director of the Office of Hematology and Oncology Products in the FDA’s Center for Drug Evaluation and Research, pointed out that in less than a year, "We have seen considerable progress in the availability of treatments for chronic lymphocytic leukemia." The other treatments for CLL are obinutuzumab (Gazyva), approved in November 2013; ibrutinib (Imbruvica), approved in February 2014; and ofatumumab (Arzerra) approved in April 2014.

Prescribing information is available at the FDA website.

[email protected]

The oral kinase inhibitor idelalisib has been approved for treating patients with relapsed chronic lymphocytic leukemia, follicular lymphoma, and small lymphocytic lymphoma, with a boxed warning about fatal and serious toxicities associated with treatment, the Food and Drug Administration announced on July 23.

The three approved indications for idelalisib – administered at a recommended starting dose of 150 mg, twice a day – are for:

 

• Relapsed chronic lymphocytic leukemia (CLL), in combination with rituximab, in patients for whom rituximab alone would be considered appropriate therapy due to other comorbidities. This is a traditional approval based on progression-free survival (PFS) data.

• Relapsed follicular B-cell non-Hodgkin’s lymphoma (FL) in patients who have received at least two prior systemic therapies.

• Relapsed small lymphocytic lymphoma (SLL) in patients who have received at least two prior systemic therapies.

The FL and SLL indications, based on objective response rates in one study, are accelerated approvals, which provides patients with serious or life-threatening diseases earlier access to a promising drug based on an effect on a surrogate endpoint considered " reasonably likely to predict clinical benefit," according to the FDA. Full approval is contingent on the manufacturer conducting trials confirming clinical benefits; if they do not, the FDA can withdraw the approval.

Idelalisib is being marketed as Zydelig, by Gilead Sciences. There is also a Risk Evaluation and Mitigation Strategy (REMS) in place for the drug, which includes a communication plan to ensure that prescribers are fully informed about treatment-associated risks. The boxed warning lists the risks of fatal and/or serious hepatoxicity (affecting 14% of treated patients); fatal and/or serious and severe diarrhea or colitis (also affecting 14% of treated patients); as well as fatal and serious pneumonitis; and fatal and serious intestinal perforation.

Idelalisib is an oral inhibitor of phosphoinositide 3-kinase (PI3K) delta, "a protein that plays a role in the activation, proliferation, and viability of B cells," according to a Gilead statement announcing the approval, which added that PI3K delta signaling "is active in many B-cell leukemias and lymphomas, and by inhibiting the protein, Zydelig blocks several cellular signaling pathways that drive B-cell viability."

The FDA granted a full approval for the CLL indication, based on a phase III study of 220 patients, which was stopped early in October 2013 at the first-specified interim analysis. Median PFS was not reached among those randomized to on idelalisib plus rituximab, but was at least 10.7 months, and was 5.5 months among those randomized to placebo plus rituximab (N. Engl. J. Med. 2014;370:997-1007). "Results from a second interim analysis continued to show a statistically significant improvement for Zydelig and Rituxan over placebo and Rituxan," the FDA statement said.

The accelerated approvals for relapsed FL and relapsed SLL were based on a single-arm phase II study of 123 patients refractory to rituximab and chemotherapy including alkylating agents, treated with idelalisib. The objective response rates were 54% among those with relapsed FL, and 58% of those with SLL in the study (N. Engl. J. Med. 2014;370:1008-18).

Common adverse events associated with treatment included diarrhea, fever, fatigue, nausea, cough, pneumonia, abdominal pain, chills, and rash; and common lab abnormalities associated with treatment included neutropenia, hypertriglyceridemia, hyperglycemia, and liver enzyme elevations, according to the FDA.

In the FDA statement, Dr. Richard Pazdur, director of the Office of Hematology and Oncology Products in the FDA’s Center for Drug Evaluation and Research, pointed out that in less than a year, "We have seen considerable progress in the availability of treatments for chronic lymphocytic leukemia." The other treatments for CLL are obinutuzumab (Gazyva), approved in November 2013; ibrutinib (Imbruvica), approved in February 2014; and ofatumumab (Arzerra) approved in April 2014.

Prescribing information is available at the FDA website.

[email protected]

Publications
Publications
Topics
Article Type
Display Headline
FDA approves idelalisib for three leukemia and lymphoma indications
Display Headline
FDA approves idelalisib for three leukemia and lymphoma indications
Legacy Keywords
oral kinase inhibitor, idelalisib, relapsed chronic lymphocytic leukemia, follicular lymphoma, small lymphocytic lymphoma, Food and Drug Administration,
Legacy Keywords
oral kinase inhibitor, idelalisib, relapsed chronic lymphocytic leukemia, follicular lymphoma, small lymphocytic lymphoma, Food and Drug Administration,
PURLs Copyright

Disallow All Ads
Alternative CME
Use ProPublica

Combo appears safe and active in CLL, NHL

Article Type
Changed
Fri, 12/16/2022 - 12:26
Display Headline
Combo appears safe and active in CLL, NHL

 

 

 

Monoclonal antibodies

Credit: Linda Bartlett

 

KOHALA COAST, HAWAII—Early results of a small, phase 1 study suggest a novel combination treatment is active and generally well-tolerated in relapsed or refractory patients with chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL) or non-Hodgkin lymphomas (NHLs).

 

The treatment consists of ublituximab (TG-1101), a monoclonal antibody that targets a unique epitope on the CD20 antigen, and TGR-1202, a next-generation PI3K delta inhibitor.

 

The combination appeared to be well tolerated overall, although infusion-related reactions were common, and nearly a quarter of patients experienced grade 3/4 neutropenia.

 

Four of 5 CLL/SLL patients experienced a partial response (PR), and the remaining patient had stable disease (SD). Among the 10 NHL patients, 1 had progressive disease, 7 had SD, and 2 achieved a PR.

 

Matthew Lunning, DO, of the University of Nebraska Medical Center in Omaha, and his colleagues presented these results in a poster at the 2014 Pan Pacific Lymphoma Conference.

 

The study is sponsored by TG Therapeutics, the company developing both drugs.

 

The researchers presented results from 8 patients with CLL/SLL, 7 patients with diffuse large B-cell lymphoma (DLBCL), 5 with follicular lymphoma (FL), and 1 patient with Richter’s syndrome (RS).

 

Patients had a median age of 64 years (range, 35-82), and they had received a median of 3 prior therapies (range, 1-9). Fifty-seven percent of patients had received 3 or more prior therapies, and 38% were refractory to their prior therapy.

 

The patients received escalating doses of TGR-1202, with a fixed dose of ublituximab—900 mg for patients with NHL and 600 mg for patients with CLL.

 

As of the data cutoff, all 21 patients were evaluable for safety, but only 15 were evaluable for efficacy.

 

Adverse events

 

The most common adverse event was infusion-related reactions, which occurred in 48% of patients. All of these events were manageable without dose reductions, and all but 1 event was grade 1 or 2 in severity.

 

Neutropenia was also common, occurring in 38% of patients. Grade 3/4 neutropenia occurred in 24% of patients. One CLL patient required a dose delay for neutropenia in cycle 1, which met the criteria for a dose-limiting toxicity.

 

No additional dose-limiting toxicities have been observed to date. Likewise, none of the patients has required dose reductions for either drug, and there were no drug-related AST/ALT elevations.

 

On the other hand, 1 patient did come off the study due to grade 1 itching that was possibly related to TGR-1202.

 

Other common adverse events associated with treatment included diarrhea (29%), nausea (29%), hoarseness (10%), muscle aches (10%), and fatigue (10%).

 

Activity in CLL/SLL

 

Of the 8 CLL/SLL patients enrolled to date, 5 were evaluable for efficacy. Four patients achieved a PR at the first efficacy assessment. The remaining patient, a CLL patient with both 17p and 11q del, achieved SD with a 44% nodal reduction at the first assessment.

 

All 5 patients achieved a greater-than-50% reduction in ALC by the first efficacy assessment. One patient achieved complete normalization of ALC (less than 4000/uL), and the other 4 patients achieved at least an 80% reduction by the first efficacy assessment.

 

The lymphocytosis generally observed in CLL patients treated with TGR-1202, similar to other PI3K delta and BTK inhibitors, appears to be mitigated by the addition of ublituximab.

 

Activity in NHL

 

Of the 13 patients in this group, 10 were evaluable for efficacy, including 5 with DLBCL, 4 with FL, and 1 with RS. Results were not as favorable in this group as they were among CLL/SLL patients, but, as the researchers pointed out, these patients were heavily pretreated.

 

 

 

Among the DLBCL patients, 2 achieved PRs with TGR-1202 and ublituximab. Both of these responses occurred at the higher dose of TGR-1202.

 

Two DLBCL patients had SD, and 1 patient progressed. DLBCL patients had a median of 3 prior treatment lines, and 3 patients had GCB DLBCL, with 1 patient classified as triple-hit lymphoma (overexpression of BCL2, BCL6, and MYC rearrangements).

 

In the FL group, all 4 patients had SD after treatment and exhibited a reduction in tumor mass at the first assessment. These patients had advanced disease and a median of 6 prior lines of therapy.

 

The RS patient also had SD following TGR-1202 and ublituximab.

 

“We have been very impressed with the safety profile and the level of activity observed to date in all patient groups with TGR-1202 in combination with ublituximab, particularly given the advanced stage of disease . . . ,” said Susan O’Brien, MD, a professor at MD Anderson Cancer Center in Houston and study chair for the CLL patient group.

 

“Of particular interest is the absence of observed elevations in AST/ALT with TGR-1202, which is a known adverse event associated with other PI3K delta inhibitors. We look forward to continuing enrollment at all trial centers of this exciting combination and presenting data on more patients at upcoming medical meetings.”

Publications
Topics

 

 

 

Monoclonal antibodies

Credit: Linda Bartlett

 

KOHALA COAST, HAWAII—Early results of a small, phase 1 study suggest a novel combination treatment is active and generally well-tolerated in relapsed or refractory patients with chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL) or non-Hodgkin lymphomas (NHLs).

 

The treatment consists of ublituximab (TG-1101), a monoclonal antibody that targets a unique epitope on the CD20 antigen, and TGR-1202, a next-generation PI3K delta inhibitor.

 

The combination appeared to be well tolerated overall, although infusion-related reactions were common, and nearly a quarter of patients experienced grade 3/4 neutropenia.

 

Four of 5 CLL/SLL patients experienced a partial response (PR), and the remaining patient had stable disease (SD). Among the 10 NHL patients, 1 had progressive disease, 7 had SD, and 2 achieved a PR.

 

Matthew Lunning, DO, of the University of Nebraska Medical Center in Omaha, and his colleagues presented these results in a poster at the 2014 Pan Pacific Lymphoma Conference.

 

The study is sponsored by TG Therapeutics, the company developing both drugs.

 

The researchers presented results from 8 patients with CLL/SLL, 7 patients with diffuse large B-cell lymphoma (DLBCL), 5 with follicular lymphoma (FL), and 1 patient with Richter’s syndrome (RS).

 

Patients had a median age of 64 years (range, 35-82), and they had received a median of 3 prior therapies (range, 1-9). Fifty-seven percent of patients had received 3 or more prior therapies, and 38% were refractory to their prior therapy.

 

The patients received escalating doses of TGR-1202, with a fixed dose of ublituximab—900 mg for patients with NHL and 600 mg for patients with CLL.

 

As of the data cutoff, all 21 patients were evaluable for safety, but only 15 were evaluable for efficacy.

 

Adverse events

 

The most common adverse event was infusion-related reactions, which occurred in 48% of patients. All of these events were manageable without dose reductions, and all but 1 event was grade 1 or 2 in severity.

 

Neutropenia was also common, occurring in 38% of patients. Grade 3/4 neutropenia occurred in 24% of patients. One CLL patient required a dose delay for neutropenia in cycle 1, which met the criteria for a dose-limiting toxicity.

 

No additional dose-limiting toxicities have been observed to date. Likewise, none of the patients has required dose reductions for either drug, and there were no drug-related AST/ALT elevations.

 

On the other hand, 1 patient did come off the study due to grade 1 itching that was possibly related to TGR-1202.

 

Other common adverse events associated with treatment included diarrhea (29%), nausea (29%), hoarseness (10%), muscle aches (10%), and fatigue (10%).

 

Activity in CLL/SLL

 

Of the 8 CLL/SLL patients enrolled to date, 5 were evaluable for efficacy. Four patients achieved a PR at the first efficacy assessment. The remaining patient, a CLL patient with both 17p and 11q del, achieved SD with a 44% nodal reduction at the first assessment.

 

All 5 patients achieved a greater-than-50% reduction in ALC by the first efficacy assessment. One patient achieved complete normalization of ALC (less than 4000/uL), and the other 4 patients achieved at least an 80% reduction by the first efficacy assessment.

 

The lymphocytosis generally observed in CLL patients treated with TGR-1202, similar to other PI3K delta and BTK inhibitors, appears to be mitigated by the addition of ublituximab.

 

Activity in NHL

 

Of the 13 patients in this group, 10 were evaluable for efficacy, including 5 with DLBCL, 4 with FL, and 1 with RS. Results were not as favorable in this group as they were among CLL/SLL patients, but, as the researchers pointed out, these patients were heavily pretreated.

 

 

 

Among the DLBCL patients, 2 achieved PRs with TGR-1202 and ublituximab. Both of these responses occurred at the higher dose of TGR-1202.

 

Two DLBCL patients had SD, and 1 patient progressed. DLBCL patients had a median of 3 prior treatment lines, and 3 patients had GCB DLBCL, with 1 patient classified as triple-hit lymphoma (overexpression of BCL2, BCL6, and MYC rearrangements).

 

In the FL group, all 4 patients had SD after treatment and exhibited a reduction in tumor mass at the first assessment. These patients had advanced disease and a median of 6 prior lines of therapy.

 

The RS patient also had SD following TGR-1202 and ublituximab.

 

“We have been very impressed with the safety profile and the level of activity observed to date in all patient groups with TGR-1202 in combination with ublituximab, particularly given the advanced stage of disease . . . ,” said Susan O’Brien, MD, a professor at MD Anderson Cancer Center in Houston and study chair for the CLL patient group.

 

“Of particular interest is the absence of observed elevations in AST/ALT with TGR-1202, which is a known adverse event associated with other PI3K delta inhibitors. We look forward to continuing enrollment at all trial centers of this exciting combination and presenting data on more patients at upcoming medical meetings.”

 

 

 

Monoclonal antibodies

Credit: Linda Bartlett

 

KOHALA COAST, HAWAII—Early results of a small, phase 1 study suggest a novel combination treatment is active and generally well-tolerated in relapsed or refractory patients with chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL) or non-Hodgkin lymphomas (NHLs).

 

The treatment consists of ublituximab (TG-1101), a monoclonal antibody that targets a unique epitope on the CD20 antigen, and TGR-1202, a next-generation PI3K delta inhibitor.

 

The combination appeared to be well tolerated overall, although infusion-related reactions were common, and nearly a quarter of patients experienced grade 3/4 neutropenia.

 

Four of 5 CLL/SLL patients experienced a partial response (PR), and the remaining patient had stable disease (SD). Among the 10 NHL patients, 1 had progressive disease, 7 had SD, and 2 achieved a PR.

 

Matthew Lunning, DO, of the University of Nebraska Medical Center in Omaha, and his colleagues presented these results in a poster at the 2014 Pan Pacific Lymphoma Conference.

 

The study is sponsored by TG Therapeutics, the company developing both drugs.

 

The researchers presented results from 8 patients with CLL/SLL, 7 patients with diffuse large B-cell lymphoma (DLBCL), 5 with follicular lymphoma (FL), and 1 patient with Richter’s syndrome (RS).

 

Patients had a median age of 64 years (range, 35-82), and they had received a median of 3 prior therapies (range, 1-9). Fifty-seven percent of patients had received 3 or more prior therapies, and 38% were refractory to their prior therapy.

 

The patients received escalating doses of TGR-1202, with a fixed dose of ublituximab—900 mg for patients with NHL and 600 mg for patients with CLL.

 

As of the data cutoff, all 21 patients were evaluable for safety, but only 15 were evaluable for efficacy.

 

Adverse events

 

The most common adverse event was infusion-related reactions, which occurred in 48% of patients. All of these events were manageable without dose reductions, and all but 1 event was grade 1 or 2 in severity.

 

Neutropenia was also common, occurring in 38% of patients. Grade 3/4 neutropenia occurred in 24% of patients. One CLL patient required a dose delay for neutropenia in cycle 1, which met the criteria for a dose-limiting toxicity.

 

No additional dose-limiting toxicities have been observed to date. Likewise, none of the patients has required dose reductions for either drug, and there were no drug-related AST/ALT elevations.

 

On the other hand, 1 patient did come off the study due to grade 1 itching that was possibly related to TGR-1202.

 

Other common adverse events associated with treatment included diarrhea (29%), nausea (29%), hoarseness (10%), muscle aches (10%), and fatigue (10%).

 

Activity in CLL/SLL

 

Of the 8 CLL/SLL patients enrolled to date, 5 were evaluable for efficacy. Four patients achieved a PR at the first efficacy assessment. The remaining patient, a CLL patient with both 17p and 11q del, achieved SD with a 44% nodal reduction at the first assessment.

 

All 5 patients achieved a greater-than-50% reduction in ALC by the first efficacy assessment. One patient achieved complete normalization of ALC (less than 4000/uL), and the other 4 patients achieved at least an 80% reduction by the first efficacy assessment.

 

The lymphocytosis generally observed in CLL patients treated with TGR-1202, similar to other PI3K delta and BTK inhibitors, appears to be mitigated by the addition of ublituximab.

 

Activity in NHL

 

Of the 13 patients in this group, 10 were evaluable for efficacy, including 5 with DLBCL, 4 with FL, and 1 with RS. Results were not as favorable in this group as they were among CLL/SLL patients, but, as the researchers pointed out, these patients were heavily pretreated.

 

 

 

Among the DLBCL patients, 2 achieved PRs with TGR-1202 and ublituximab. Both of these responses occurred at the higher dose of TGR-1202.

 

Two DLBCL patients had SD, and 1 patient progressed. DLBCL patients had a median of 3 prior treatment lines, and 3 patients had GCB DLBCL, with 1 patient classified as triple-hit lymphoma (overexpression of BCL2, BCL6, and MYC rearrangements).

 

In the FL group, all 4 patients had SD after treatment and exhibited a reduction in tumor mass at the first assessment. These patients had advanced disease and a median of 6 prior lines of therapy.

 

The RS patient also had SD following TGR-1202 and ublituximab.

 

“We have been very impressed with the safety profile and the level of activity observed to date in all patient groups with TGR-1202 in combination with ublituximab, particularly given the advanced stage of disease . . . ,” said Susan O’Brien, MD, a professor at MD Anderson Cancer Center in Houston and study chair for the CLL patient group.

 

“Of particular interest is the absence of observed elevations in AST/ALT with TGR-1202, which is a known adverse event associated with other PI3K delta inhibitors. We look forward to continuing enrollment at all trial centers of this exciting combination and presenting data on more patients at upcoming medical meetings.”

Publications
Publications
Topics
Article Type
Display Headline
Combo appears safe and active in CLL, NHL
Display Headline
Combo appears safe and active in CLL, NHL
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

Fasting can have beneficial effects in cancer setting

Article Type
Changed
Wed, 07/23/2014 - 06:00
Display Headline
Fasting can have beneficial effects in cancer setting

Hematopoietic stem cells

in the bone marrow

New research indicates that cycles of prolonged fasting may prevent chemotherapy-induced immunosuppressive toxicity and induce regeneration of the hematopoietic system.

Long periods of fasting reduced damage in bone marrow stem and progenitor cells and protected both mice and humans from chemotoxicity.

In mice, the fasting cycles “flipped a regenerative switch,” changing the signaling pathways for hematopoietic stem cells (HSCs).

Researchers reported these results in Cell Stem Cell.

“We could not predict that prolonged fasting would have such a remarkable effect in promoting stem cell-based regeneration of the hematopoietic system,” said study author Valter Longo, PhD, of the University of Southern California in Los Angeles.

“When you starve, the system tries to save energy, and one of the things it can do to save energy is to recycle a lot of the immune cells that are not needed, especially those that may be damaged. What we started noticing in both our human work and animal work is that the white blood cell count goes down with prolonged fasting. Then, when you re-feed, the blood cells come back. So we started thinking, well, where does it come from?”

The researchers found that prolonged fasting reduced the enzyme PKA, which regulates HSC self-renewal and pluripotency.

“PKA is the key gene that needs to shut down in order for these stem cells to switch into regenerative mode,” Dr Longo said. “It gives the ‘okay’ for stem cells to go ahead and begin proliferating and rebuild the entire system.”

Prolonged fasting also lowered levels of IGF-1, a growth-factor hormone that has been linked to aging, tumor progression, and cancer risk.

In addition to downregulating the IGF-1/PKA pathway in HSCs, prolonged fasting protected hematopoietic cells from chemotoxicity and promoted HSC self-renewal to reverse immunosuppression.

Experiments revealed that inhibiting IGF-1 or PKA signaling mimicked the effects of prolonged fasting.

The researchers also analyzed a small group of patients from a pilot study evaluating the effects of fasting before chemotherapy. Fasting for 72 hours, but not 24 hours, ensured that patients had normal lymphocyte counts and maintained a normal lineage balance in white blood cells after chemotherapy.

Dr Longo’s lab is now conducting further research on controlled dietary interventions and stem cell regeneration in both animal and clinical studies.

Publications
Topics

Hematopoietic stem cells

in the bone marrow

New research indicates that cycles of prolonged fasting may prevent chemotherapy-induced immunosuppressive toxicity and induce regeneration of the hematopoietic system.

Long periods of fasting reduced damage in bone marrow stem and progenitor cells and protected both mice and humans from chemotoxicity.

In mice, the fasting cycles “flipped a regenerative switch,” changing the signaling pathways for hematopoietic stem cells (HSCs).

Researchers reported these results in Cell Stem Cell.

“We could not predict that prolonged fasting would have such a remarkable effect in promoting stem cell-based regeneration of the hematopoietic system,” said study author Valter Longo, PhD, of the University of Southern California in Los Angeles.

“When you starve, the system tries to save energy, and one of the things it can do to save energy is to recycle a lot of the immune cells that are not needed, especially those that may be damaged. What we started noticing in both our human work and animal work is that the white blood cell count goes down with prolonged fasting. Then, when you re-feed, the blood cells come back. So we started thinking, well, where does it come from?”

The researchers found that prolonged fasting reduced the enzyme PKA, which regulates HSC self-renewal and pluripotency.

“PKA is the key gene that needs to shut down in order for these stem cells to switch into regenerative mode,” Dr Longo said. “It gives the ‘okay’ for stem cells to go ahead and begin proliferating and rebuild the entire system.”

Prolonged fasting also lowered levels of IGF-1, a growth-factor hormone that has been linked to aging, tumor progression, and cancer risk.

In addition to downregulating the IGF-1/PKA pathway in HSCs, prolonged fasting protected hematopoietic cells from chemotoxicity and promoted HSC self-renewal to reverse immunosuppression.

Experiments revealed that inhibiting IGF-1 or PKA signaling mimicked the effects of prolonged fasting.

The researchers also analyzed a small group of patients from a pilot study evaluating the effects of fasting before chemotherapy. Fasting for 72 hours, but not 24 hours, ensured that patients had normal lymphocyte counts and maintained a normal lineage balance in white blood cells after chemotherapy.

Dr Longo’s lab is now conducting further research on controlled dietary interventions and stem cell regeneration in both animal and clinical studies.

Hematopoietic stem cells

in the bone marrow

New research indicates that cycles of prolonged fasting may prevent chemotherapy-induced immunosuppressive toxicity and induce regeneration of the hematopoietic system.

Long periods of fasting reduced damage in bone marrow stem and progenitor cells and protected both mice and humans from chemotoxicity.

In mice, the fasting cycles “flipped a regenerative switch,” changing the signaling pathways for hematopoietic stem cells (HSCs).

Researchers reported these results in Cell Stem Cell.

“We could not predict that prolonged fasting would have such a remarkable effect in promoting stem cell-based regeneration of the hematopoietic system,” said study author Valter Longo, PhD, of the University of Southern California in Los Angeles.

“When you starve, the system tries to save energy, and one of the things it can do to save energy is to recycle a lot of the immune cells that are not needed, especially those that may be damaged. What we started noticing in both our human work and animal work is that the white blood cell count goes down with prolonged fasting. Then, when you re-feed, the blood cells come back. So we started thinking, well, where does it come from?”

The researchers found that prolonged fasting reduced the enzyme PKA, which regulates HSC self-renewal and pluripotency.

“PKA is the key gene that needs to shut down in order for these stem cells to switch into regenerative mode,” Dr Longo said. “It gives the ‘okay’ for stem cells to go ahead and begin proliferating and rebuild the entire system.”

Prolonged fasting also lowered levels of IGF-1, a growth-factor hormone that has been linked to aging, tumor progression, and cancer risk.

In addition to downregulating the IGF-1/PKA pathway in HSCs, prolonged fasting protected hematopoietic cells from chemotoxicity and promoted HSC self-renewal to reverse immunosuppression.

Experiments revealed that inhibiting IGF-1 or PKA signaling mimicked the effects of prolonged fasting.

The researchers also analyzed a small group of patients from a pilot study evaluating the effects of fasting before chemotherapy. Fasting for 72 hours, but not 24 hours, ensured that patients had normal lymphocyte counts and maintained a normal lineage balance in white blood cells after chemotherapy.

Dr Longo’s lab is now conducting further research on controlled dietary interventions and stem cell regeneration in both animal and clinical studies.

Publications
Publications
Topics
Article Type
Display Headline
Fasting can have beneficial effects in cancer setting
Display Headline
Fasting can have beneficial effects in cancer setting
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

Drug approved to treat NHL in Israel

Article Type
Changed
Wed, 07/23/2014 - 05:00
Display Headline
Drug approved to treat NHL in Israel

Diffuse large B-cell lymphoma

The Israeli Ministry of Health has granted approval for the antineoplastic agent pixantrone (Pixuvri).

The drug is now approved as monotherapy for adults with relapsed or refractory aggressive B-cell non-Hodgkin lymphoma (NHL) who have received fewer than 4 previous courses of treatment.

The benefit of pixantrone has not been established when used as fifth-line or greater treatment in patients who were refractory to their last therapy.

Pixantrone will be distributed in Israel by the Neopharm Group.

“The approval of Pixuvri in Israel provides patients with aggressive B-cell NHL who have failed second- or third-line therapy a new approved option, where none existed before, that can effectively treat their disease with manageable side effects,” said Abraham Avigdor, MD, of Tel Aviv University.

“Patients who have relapsed after second-line therapy have a poor survival outcome. It is vital to have additional treatment options available, like Pixuvri, so we can provide these patients the best care possible and help them battle their disease.”

The main study of pixantrone, the phase 3 EXTEND PIX301 trial, compared the drug to other chemotherapeutic agents in patients with relapsed or refractory NHL. The response rate was 20% in the pixantrone arm and 6% in the comparator arm.

In addition, patients receiving pixantrone had longer progression-free survival than patients in the comparator group, with a median of 10.2 months and 7.6 months, respectively.

However, grade 3/4 adverse events—including neutropenia, leukopenia, and thrombocytopenia—were more common in the pixantrone arm.

Pixantrone is already marketed in the European Union. In 2012, the European Commission granted conditional marketing authorization for the drug as monotherapy for adults with relapsed or refractory aggressive B-cell NHL.

Under the provisions of the conditional marketing authorization, Cell Therapeutics, Inc., the company developing pixantrone, will be required to complete a post-marketing study aimed at confirming the drug’s clinical benefit.

The European Medicines Agency’s Committee for Medicinal Products for Human Use has accepted PIX306, a randomized, phase 3 trial comparing pixantrone plus rituximab to gemcitabine plus rituximab in patients who have relapsed after 1 to 3 prior regimens for aggressive B-cell NHL and who are not eligible for autologous stem cell transplant.

As a condition of approval, Cell Therapeutics has agreed to have the trial data available by June 2015.

Pixantrone is not approved for use in the US.

Publications
Topics

Diffuse large B-cell lymphoma

The Israeli Ministry of Health has granted approval for the antineoplastic agent pixantrone (Pixuvri).

The drug is now approved as monotherapy for adults with relapsed or refractory aggressive B-cell non-Hodgkin lymphoma (NHL) who have received fewer than 4 previous courses of treatment.

The benefit of pixantrone has not been established when used as fifth-line or greater treatment in patients who were refractory to their last therapy.

Pixantrone will be distributed in Israel by the Neopharm Group.

“The approval of Pixuvri in Israel provides patients with aggressive B-cell NHL who have failed second- or third-line therapy a new approved option, where none existed before, that can effectively treat their disease with manageable side effects,” said Abraham Avigdor, MD, of Tel Aviv University.

“Patients who have relapsed after second-line therapy have a poor survival outcome. It is vital to have additional treatment options available, like Pixuvri, so we can provide these patients the best care possible and help them battle their disease.”

The main study of pixantrone, the phase 3 EXTEND PIX301 trial, compared the drug to other chemotherapeutic agents in patients with relapsed or refractory NHL. The response rate was 20% in the pixantrone arm and 6% in the comparator arm.

In addition, patients receiving pixantrone had longer progression-free survival than patients in the comparator group, with a median of 10.2 months and 7.6 months, respectively.

However, grade 3/4 adverse events—including neutropenia, leukopenia, and thrombocytopenia—were more common in the pixantrone arm.

Pixantrone is already marketed in the European Union. In 2012, the European Commission granted conditional marketing authorization for the drug as monotherapy for adults with relapsed or refractory aggressive B-cell NHL.

Under the provisions of the conditional marketing authorization, Cell Therapeutics, Inc., the company developing pixantrone, will be required to complete a post-marketing study aimed at confirming the drug’s clinical benefit.

The European Medicines Agency’s Committee for Medicinal Products for Human Use has accepted PIX306, a randomized, phase 3 trial comparing pixantrone plus rituximab to gemcitabine plus rituximab in patients who have relapsed after 1 to 3 prior regimens for aggressive B-cell NHL and who are not eligible for autologous stem cell transplant.

As a condition of approval, Cell Therapeutics has agreed to have the trial data available by June 2015.

Pixantrone is not approved for use in the US.

Diffuse large B-cell lymphoma

The Israeli Ministry of Health has granted approval for the antineoplastic agent pixantrone (Pixuvri).

The drug is now approved as monotherapy for adults with relapsed or refractory aggressive B-cell non-Hodgkin lymphoma (NHL) who have received fewer than 4 previous courses of treatment.

The benefit of pixantrone has not been established when used as fifth-line or greater treatment in patients who were refractory to their last therapy.

Pixantrone will be distributed in Israel by the Neopharm Group.

“The approval of Pixuvri in Israel provides patients with aggressive B-cell NHL who have failed second- or third-line therapy a new approved option, where none existed before, that can effectively treat their disease with manageable side effects,” said Abraham Avigdor, MD, of Tel Aviv University.

“Patients who have relapsed after second-line therapy have a poor survival outcome. It is vital to have additional treatment options available, like Pixuvri, so we can provide these patients the best care possible and help them battle their disease.”

The main study of pixantrone, the phase 3 EXTEND PIX301 trial, compared the drug to other chemotherapeutic agents in patients with relapsed or refractory NHL. The response rate was 20% in the pixantrone arm and 6% in the comparator arm.

In addition, patients receiving pixantrone had longer progression-free survival than patients in the comparator group, with a median of 10.2 months and 7.6 months, respectively.

However, grade 3/4 adverse events—including neutropenia, leukopenia, and thrombocytopenia—were more common in the pixantrone arm.

Pixantrone is already marketed in the European Union. In 2012, the European Commission granted conditional marketing authorization for the drug as monotherapy for adults with relapsed or refractory aggressive B-cell NHL.

Under the provisions of the conditional marketing authorization, Cell Therapeutics, Inc., the company developing pixantrone, will be required to complete a post-marketing study aimed at confirming the drug’s clinical benefit.

The European Medicines Agency’s Committee for Medicinal Products for Human Use has accepted PIX306, a randomized, phase 3 trial comparing pixantrone plus rituximab to gemcitabine plus rituximab in patients who have relapsed after 1 to 3 prior regimens for aggressive B-cell NHL and who are not eligible for autologous stem cell transplant.

As a condition of approval, Cell Therapeutics has agreed to have the trial data available by June 2015.

Pixantrone is not approved for use in the US.

Publications
Publications
Topics
Article Type
Display Headline
Drug approved to treat NHL in Israel
Display Headline
Drug approved to treat NHL in Israel
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

Protein map may point to new cancer treatments

Article Type
Changed
Tue, 07/22/2014 - 05:00
Display Headline
Protein map may point to new cancer treatments

Image showing mitosis, with

the endoplasmic reticulum

in green, mitochondria in red,

and chromosomes in blue

Credit: Wellcome Images

Researchers say they’ve uncovered the structure of one of the most important protein complexes involved in cell division, and their finding has implications for cancer treatment.

The team mapped the anaphase-promoting complex (APC/C), which performs a wide range of tasks associated with mitosis.

The researchers believe that seeing APC/C in unprecedented detail could transform our understanding of how cells divide and reveal binding sites for future cancer drugs.

“It’s very rewarding to finally tie down the detailed structure of this important protein, which is both one of the most important and most complicated found in all of nature,” said David Barford, DPhil, of the Medical Research Council Laboratory of Molecular Biology in Cambridge, UK.

“We hope our discovery will open up whole new avenues of research that increase our understanding of the process of mitosis and ultimately lead to the discovery of new cancer drugs.”

Dr Barford and his colleagues detailed their discovery in Nature.

The researchers reconstituted human APC/C and used a combination of electron microscopy and imaging software to visualize it at a resolution of less than a billionth of a meter.

The resolution was so fine that it allowed the team to see the secondary structure—the set of basic building blocks that combine to form every protein. Alpha-helix rods and folded beta-sheet constructions were clearly visible within the 20 subunits of the APC/C, defining the overall architecture of the complex.

Previous studies led by the same team had shown a globular structure for APC/C in much lower resolution, but the secondary structure had not been mapped.

Each of the APC/C’s subunits bond and mesh with other units at different points in the cell cycle, allowing it to control a range of mitotic processes, including the initiation of DNA replication, the segregation of chromosomes along spindles, and cytokinesis. Disrupting each of these processes could selectively kill cancer cells or prevent them from dividing.

“The fantastic insights into molecular structure provided by this study are a vivid illustration of the critical role played by fundamental cell biology in cancer research,” said Paul Workman, PhD, of The Institute of Cancer Research, London.

“The new study is a major step forward in our understanding of cell division. When this process goes awry, it is a critical difference that separates cancer cells from their healthy counterparts. Understanding exactly how cancer cells divide inappropriately is crucial to the discovery of innovative cancer treatments to improve outcomes for cancer patients.”

Publications
Topics

Image showing mitosis, with

the endoplasmic reticulum

in green, mitochondria in red,

and chromosomes in blue

Credit: Wellcome Images

Researchers say they’ve uncovered the structure of one of the most important protein complexes involved in cell division, and their finding has implications for cancer treatment.

The team mapped the anaphase-promoting complex (APC/C), which performs a wide range of tasks associated with mitosis.

The researchers believe that seeing APC/C in unprecedented detail could transform our understanding of how cells divide and reveal binding sites for future cancer drugs.

“It’s very rewarding to finally tie down the detailed structure of this important protein, which is both one of the most important and most complicated found in all of nature,” said David Barford, DPhil, of the Medical Research Council Laboratory of Molecular Biology in Cambridge, UK.

“We hope our discovery will open up whole new avenues of research that increase our understanding of the process of mitosis and ultimately lead to the discovery of new cancer drugs.”

Dr Barford and his colleagues detailed their discovery in Nature.

The researchers reconstituted human APC/C and used a combination of electron microscopy and imaging software to visualize it at a resolution of less than a billionth of a meter.

The resolution was so fine that it allowed the team to see the secondary structure—the set of basic building blocks that combine to form every protein. Alpha-helix rods and folded beta-sheet constructions were clearly visible within the 20 subunits of the APC/C, defining the overall architecture of the complex.

Previous studies led by the same team had shown a globular structure for APC/C in much lower resolution, but the secondary structure had not been mapped.

Each of the APC/C’s subunits bond and mesh with other units at different points in the cell cycle, allowing it to control a range of mitotic processes, including the initiation of DNA replication, the segregation of chromosomes along spindles, and cytokinesis. Disrupting each of these processes could selectively kill cancer cells or prevent them from dividing.

“The fantastic insights into molecular structure provided by this study are a vivid illustration of the critical role played by fundamental cell biology in cancer research,” said Paul Workman, PhD, of The Institute of Cancer Research, London.

“The new study is a major step forward in our understanding of cell division. When this process goes awry, it is a critical difference that separates cancer cells from their healthy counterparts. Understanding exactly how cancer cells divide inappropriately is crucial to the discovery of innovative cancer treatments to improve outcomes for cancer patients.”

Image showing mitosis, with

the endoplasmic reticulum

in green, mitochondria in red,

and chromosomes in blue

Credit: Wellcome Images

Researchers say they’ve uncovered the structure of one of the most important protein complexes involved in cell division, and their finding has implications for cancer treatment.

The team mapped the anaphase-promoting complex (APC/C), which performs a wide range of tasks associated with mitosis.

The researchers believe that seeing APC/C in unprecedented detail could transform our understanding of how cells divide and reveal binding sites for future cancer drugs.

“It’s very rewarding to finally tie down the detailed structure of this important protein, which is both one of the most important and most complicated found in all of nature,” said David Barford, DPhil, of the Medical Research Council Laboratory of Molecular Biology in Cambridge, UK.

“We hope our discovery will open up whole new avenues of research that increase our understanding of the process of mitosis and ultimately lead to the discovery of new cancer drugs.”

Dr Barford and his colleagues detailed their discovery in Nature.

The researchers reconstituted human APC/C and used a combination of electron microscopy and imaging software to visualize it at a resolution of less than a billionth of a meter.

The resolution was so fine that it allowed the team to see the secondary structure—the set of basic building blocks that combine to form every protein. Alpha-helix rods and folded beta-sheet constructions were clearly visible within the 20 subunits of the APC/C, defining the overall architecture of the complex.

Previous studies led by the same team had shown a globular structure for APC/C in much lower resolution, but the secondary structure had not been mapped.

Each of the APC/C’s subunits bond and mesh with other units at different points in the cell cycle, allowing it to control a range of mitotic processes, including the initiation of DNA replication, the segregation of chromosomes along spindles, and cytokinesis. Disrupting each of these processes could selectively kill cancer cells or prevent them from dividing.

“The fantastic insights into molecular structure provided by this study are a vivid illustration of the critical role played by fundamental cell biology in cancer research,” said Paul Workman, PhD, of The Institute of Cancer Research, London.

“The new study is a major step forward in our understanding of cell division. When this process goes awry, it is a critical difference that separates cancer cells from their healthy counterparts. Understanding exactly how cancer cells divide inappropriately is crucial to the discovery of innovative cancer treatments to improve outcomes for cancer patients.”

Publications
Publications
Topics
Article Type
Display Headline
Protein map may point to new cancer treatments
Display Headline
Protein map may point to new cancer treatments
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

Two blood cancer patients cleared of HIV

Article Type
Changed
Mon, 07/21/2014 - 06:00
Display Headline
Two blood cancer patients cleared of HIV

HIV budding from a lymphocyte

Credit: CDC

MELBOURNE—Two men with hematologic malignancies who were also HIV-positive appear to be free of the virus after receiving stem cell transplants.

The patients have undetectable levels of HIV and remain free of their cancers—acute myeloid leukemia and non-Hodgkin lymphoma—more than 3 years after their transplants.

Importantly, the patients’ stem cell donors were not homozygous for CCR5-delta 32, a mutation that affords protection against HIV.

The researchers said these results herald a new direction in HIV research and provide hope for HIV-positive patients with leukemia and lymphoma.

The work was presented at the “Towards an HIV Cure Symposium,” which is part of the 20th International AIDS Conference.

Both patients were treated at St Vincent’s Hospital in partnership with the University of New South Wales’s Kirby Institute in Sydney, Australia.

One patient underwent a transplant in 2010 to treat his non-Hodgkin lymphoma, and his donor had 1 copy of CCR5-delta 32.

The second patient underwent a similar procedure for acute myeloid leukemia in 2011, and his donor did not have any copies of CCR5-delta 32.

Nevertheless, both patients were successfully cleared of HIV, although they remain on antiretroviral therapy as a protective measure.

“We’re so pleased that both patients are doing reasonably well years after the treatment for their cancers and remain free of both the original cancer and the HIV virus,” said David Cooper, MBBS, MD, DSc, of the Kirby Institute and St Vincent’s Hospital.

Until now, the only person considered to have cleared HIV is an American man, Timothy Ray Brown, who underwent 2 stem cell transplants in Berlin (in 2007 and 2008).

The cells in his second transplant included both copies of CCR5-delta 32, which affords protection against HIV and is found in less than 1% of the population. The man is no longer on antiretroviral therapy and remains free of HIV.

In Boston, 2 other patients underwent similar transplants in 2012, but the donor cells did not contain CCR5-delta 32. In both cases, HIV returned after antiretroviral treatment was stopped.

“It is very difficult to find a match for bone marrow donors and even more so to find one that affords protective immunity against HIV,” Dr Cooper said.

While his group’s results are a significant development, the researchers stressed that transplants are not a general functional “cure” for the up to 38.8 million people infected with HIV worldwide.

“This is a terrific, unexpected result for people with malignancy and HIV,” said Sam Milliken, MBBS, of St Vincent’s Hospital. “It may well give us a whole new insight into HIV, using the principles of stem cell transplantation.”

“It is important to caution that, at this stage, this form of treatment is far too dangerous for treating patients with HIV alone, but there may be potential for using transplants as an effective treatment modality for HIV down the track.”

The researchers said the 2 Sydney patients will be the subject of investigations to determine where any residual virus might be hiding and how it can be controlled. And the patients’ results point to a new direction for HIV research.

“We still don’t know why these patients have undetectable viral loads,” said Kersten Koelsch, MD, of the Kirby Institute. “One theory is that the induction therapy helps to destroy the cells in which the virus is hiding and that any remaining infected cells are destroyed by the patient’s new immune system.”

“We need more research to establish why and how bone marrow transplantation clears the virus. We also want to explore the predictors of sustained viral clearance and how this might be able to be exploited without the need for bone marrow transplantation.”

 

 

For the time being, the results mean that more patients who are eligible for transplant might be able to participate in clinical trials to determine the value of this procedure in HIV.

Publications
Topics

HIV budding from a lymphocyte

Credit: CDC

MELBOURNE—Two men with hematologic malignancies who were also HIV-positive appear to be free of the virus after receiving stem cell transplants.

The patients have undetectable levels of HIV and remain free of their cancers—acute myeloid leukemia and non-Hodgkin lymphoma—more than 3 years after their transplants.

Importantly, the patients’ stem cell donors were not homozygous for CCR5-delta 32, a mutation that affords protection against HIV.

The researchers said these results herald a new direction in HIV research and provide hope for HIV-positive patients with leukemia and lymphoma.

The work was presented at the “Towards an HIV Cure Symposium,” which is part of the 20th International AIDS Conference.

Both patients were treated at St Vincent’s Hospital in partnership with the University of New South Wales’s Kirby Institute in Sydney, Australia.

One patient underwent a transplant in 2010 to treat his non-Hodgkin lymphoma, and his donor had 1 copy of CCR5-delta 32.

The second patient underwent a similar procedure for acute myeloid leukemia in 2011, and his donor did not have any copies of CCR5-delta 32.

Nevertheless, both patients were successfully cleared of HIV, although they remain on antiretroviral therapy as a protective measure.

“We’re so pleased that both patients are doing reasonably well years after the treatment for their cancers and remain free of both the original cancer and the HIV virus,” said David Cooper, MBBS, MD, DSc, of the Kirby Institute and St Vincent’s Hospital.

Until now, the only person considered to have cleared HIV is an American man, Timothy Ray Brown, who underwent 2 stem cell transplants in Berlin (in 2007 and 2008).

The cells in his second transplant included both copies of CCR5-delta 32, which affords protection against HIV and is found in less than 1% of the population. The man is no longer on antiretroviral therapy and remains free of HIV.

In Boston, 2 other patients underwent similar transplants in 2012, but the donor cells did not contain CCR5-delta 32. In both cases, HIV returned after antiretroviral treatment was stopped.

“It is very difficult to find a match for bone marrow donors and even more so to find one that affords protective immunity against HIV,” Dr Cooper said.

While his group’s results are a significant development, the researchers stressed that transplants are not a general functional “cure” for the up to 38.8 million people infected with HIV worldwide.

“This is a terrific, unexpected result for people with malignancy and HIV,” said Sam Milliken, MBBS, of St Vincent’s Hospital. “It may well give us a whole new insight into HIV, using the principles of stem cell transplantation.”

“It is important to caution that, at this stage, this form of treatment is far too dangerous for treating patients with HIV alone, but there may be potential for using transplants as an effective treatment modality for HIV down the track.”

The researchers said the 2 Sydney patients will be the subject of investigations to determine where any residual virus might be hiding and how it can be controlled. And the patients’ results point to a new direction for HIV research.

“We still don’t know why these patients have undetectable viral loads,” said Kersten Koelsch, MD, of the Kirby Institute. “One theory is that the induction therapy helps to destroy the cells in which the virus is hiding and that any remaining infected cells are destroyed by the patient’s new immune system.”

“We need more research to establish why and how bone marrow transplantation clears the virus. We also want to explore the predictors of sustained viral clearance and how this might be able to be exploited without the need for bone marrow transplantation.”

 

 

For the time being, the results mean that more patients who are eligible for transplant might be able to participate in clinical trials to determine the value of this procedure in HIV.

HIV budding from a lymphocyte

Credit: CDC

MELBOURNE—Two men with hematologic malignancies who were also HIV-positive appear to be free of the virus after receiving stem cell transplants.

The patients have undetectable levels of HIV and remain free of their cancers—acute myeloid leukemia and non-Hodgkin lymphoma—more than 3 years after their transplants.

Importantly, the patients’ stem cell donors were not homozygous for CCR5-delta 32, a mutation that affords protection against HIV.

The researchers said these results herald a new direction in HIV research and provide hope for HIV-positive patients with leukemia and lymphoma.

The work was presented at the “Towards an HIV Cure Symposium,” which is part of the 20th International AIDS Conference.

Both patients were treated at St Vincent’s Hospital in partnership with the University of New South Wales’s Kirby Institute in Sydney, Australia.

One patient underwent a transplant in 2010 to treat his non-Hodgkin lymphoma, and his donor had 1 copy of CCR5-delta 32.

The second patient underwent a similar procedure for acute myeloid leukemia in 2011, and his donor did not have any copies of CCR5-delta 32.

Nevertheless, both patients were successfully cleared of HIV, although they remain on antiretroviral therapy as a protective measure.

“We’re so pleased that both patients are doing reasonably well years after the treatment for their cancers and remain free of both the original cancer and the HIV virus,” said David Cooper, MBBS, MD, DSc, of the Kirby Institute and St Vincent’s Hospital.

Until now, the only person considered to have cleared HIV is an American man, Timothy Ray Brown, who underwent 2 stem cell transplants in Berlin (in 2007 and 2008).

The cells in his second transplant included both copies of CCR5-delta 32, which affords protection against HIV and is found in less than 1% of the population. The man is no longer on antiretroviral therapy and remains free of HIV.

In Boston, 2 other patients underwent similar transplants in 2012, but the donor cells did not contain CCR5-delta 32. In both cases, HIV returned after antiretroviral treatment was stopped.

“It is very difficult to find a match for bone marrow donors and even more so to find one that affords protective immunity against HIV,” Dr Cooper said.

While his group’s results are a significant development, the researchers stressed that transplants are not a general functional “cure” for the up to 38.8 million people infected with HIV worldwide.

“This is a terrific, unexpected result for people with malignancy and HIV,” said Sam Milliken, MBBS, of St Vincent’s Hospital. “It may well give us a whole new insight into HIV, using the principles of stem cell transplantation.”

“It is important to caution that, at this stage, this form of treatment is far too dangerous for treating patients with HIV alone, but there may be potential for using transplants as an effective treatment modality for HIV down the track.”

The researchers said the 2 Sydney patients will be the subject of investigations to determine where any residual virus might be hiding and how it can be controlled. And the patients’ results point to a new direction for HIV research.

“We still don’t know why these patients have undetectable viral loads,” said Kersten Koelsch, MD, of the Kirby Institute. “One theory is that the induction therapy helps to destroy the cells in which the virus is hiding and that any remaining infected cells are destroyed by the patient’s new immune system.”

“We need more research to establish why and how bone marrow transplantation clears the virus. We also want to explore the predictors of sustained viral clearance and how this might be able to be exploited without the need for bone marrow transplantation.”

 

 

For the time being, the results mean that more patients who are eligible for transplant might be able to participate in clinical trials to determine the value of this procedure in HIV.

Publications
Publications
Topics
Article Type
Display Headline
Two blood cancer patients cleared of HIV
Display Headline
Two blood cancer patients cleared of HIV
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

Nanoparticles could improve cancer diagnosis

Article Type
Changed
Fri, 07/18/2014 - 05:00
Display Headline
Nanoparticles could improve cancer diagnosis

MRI scanner

Self-assembling nanoparticles may help physicians diagnose cancers earlier, according to a study published in Angewandte Chemie.

The nanoparticles boost the effectiveness of magnetic resonance imaging (MRI) by specifically seeking out CXCR4 receptors, which are found in cancerous cells.

The iron oxide nanoparticles are coated with peptide ligands that target tumor sites. When the particles find a tumor, they begin to interact with the cancerous cells.

Cancer-specific matrix metalloproteinase biomarkers prompt the nanoparticles to self-assemble into larger particles. And these larger particles are more visible on an MRI scan.

Researchers used cancer cells and mouse models to compare the effects of the self-assembling nanoparticles in MRI scanning against commonly used imaging agents. The nanoparticles produced a more powerful signal and created a clearer image of the tumor.

The team said the nanoparticles increase the sensitivity of MRI scans and could ultimately improve physicians’ ability to detect cancerous cells at much earlier stages of development.

“By improving the sensitivity of an MRI examination, our aim is to help doctors spot something that might be cancerous much more quickly,” said study author Nicholas Long, PhD, of Imperial College London in the UK. “This would enable patients to receive effective treatment sooner, which would hopefully improve survival rates from cancer.”

In addition to improving the sensitivity of MRI scans, the nanoparticles also appear to be safe. Before testing and injecting the particles into mice, the researchers had to ensure the particles would not become so big as to cause damage.

The team injected the particles into a saline solution inside a petri dish and monitored their growth over a 4-hour period. The nanoparticles grew from 100 nm to 800 nm, which was still small enough not to cause any harm.

Now, the researchers are working to enhance the effectiveness of the nanoparticles. And they hope to test their design in a human trial within the next 3 to 5 years.

“We would like to improve the design to make it even easier for doctors to spot a tumor and for surgeons to then operate on it,” Dr Long said. “We’re now trying to add an extra optical signal so that the nanoparticle would light up with a luminescent probe once it had found its target. So, combined with the better MRI signal, it will make it even easier to identify tumors.”

Publications
Topics

MRI scanner

Self-assembling nanoparticles may help physicians diagnose cancers earlier, according to a study published in Angewandte Chemie.

The nanoparticles boost the effectiveness of magnetic resonance imaging (MRI) by specifically seeking out CXCR4 receptors, which are found in cancerous cells.

The iron oxide nanoparticles are coated with peptide ligands that target tumor sites. When the particles find a tumor, they begin to interact with the cancerous cells.

Cancer-specific matrix metalloproteinase biomarkers prompt the nanoparticles to self-assemble into larger particles. And these larger particles are more visible on an MRI scan.

Researchers used cancer cells and mouse models to compare the effects of the self-assembling nanoparticles in MRI scanning against commonly used imaging agents. The nanoparticles produced a more powerful signal and created a clearer image of the tumor.

The team said the nanoparticles increase the sensitivity of MRI scans and could ultimately improve physicians’ ability to detect cancerous cells at much earlier stages of development.

“By improving the sensitivity of an MRI examination, our aim is to help doctors spot something that might be cancerous much more quickly,” said study author Nicholas Long, PhD, of Imperial College London in the UK. “This would enable patients to receive effective treatment sooner, which would hopefully improve survival rates from cancer.”

In addition to improving the sensitivity of MRI scans, the nanoparticles also appear to be safe. Before testing and injecting the particles into mice, the researchers had to ensure the particles would not become so big as to cause damage.

The team injected the particles into a saline solution inside a petri dish and monitored their growth over a 4-hour period. The nanoparticles grew from 100 nm to 800 nm, which was still small enough not to cause any harm.

Now, the researchers are working to enhance the effectiveness of the nanoparticles. And they hope to test their design in a human trial within the next 3 to 5 years.

“We would like to improve the design to make it even easier for doctors to spot a tumor and for surgeons to then operate on it,” Dr Long said. “We’re now trying to add an extra optical signal so that the nanoparticle would light up with a luminescent probe once it had found its target. So, combined with the better MRI signal, it will make it even easier to identify tumors.”

MRI scanner

Self-assembling nanoparticles may help physicians diagnose cancers earlier, according to a study published in Angewandte Chemie.

The nanoparticles boost the effectiveness of magnetic resonance imaging (MRI) by specifically seeking out CXCR4 receptors, which are found in cancerous cells.

The iron oxide nanoparticles are coated with peptide ligands that target tumor sites. When the particles find a tumor, they begin to interact with the cancerous cells.

Cancer-specific matrix metalloproteinase biomarkers prompt the nanoparticles to self-assemble into larger particles. And these larger particles are more visible on an MRI scan.

Researchers used cancer cells and mouse models to compare the effects of the self-assembling nanoparticles in MRI scanning against commonly used imaging agents. The nanoparticles produced a more powerful signal and created a clearer image of the tumor.

The team said the nanoparticles increase the sensitivity of MRI scans and could ultimately improve physicians’ ability to detect cancerous cells at much earlier stages of development.

“By improving the sensitivity of an MRI examination, our aim is to help doctors spot something that might be cancerous much more quickly,” said study author Nicholas Long, PhD, of Imperial College London in the UK. “This would enable patients to receive effective treatment sooner, which would hopefully improve survival rates from cancer.”

In addition to improving the sensitivity of MRI scans, the nanoparticles also appear to be safe. Before testing and injecting the particles into mice, the researchers had to ensure the particles would not become so big as to cause damage.

The team injected the particles into a saline solution inside a petri dish and monitored their growth over a 4-hour period. The nanoparticles grew from 100 nm to 800 nm, which was still small enough not to cause any harm.

Now, the researchers are working to enhance the effectiveness of the nanoparticles. And they hope to test their design in a human trial within the next 3 to 5 years.

“We would like to improve the design to make it even easier for doctors to spot a tumor and for surgeons to then operate on it,” Dr Long said. “We’re now trying to add an extra optical signal so that the nanoparticle would light up with a luminescent probe once it had found its target. So, combined with the better MRI signal, it will make it even easier to identify tumors.”

Publications
Publications
Topics
Article Type
Display Headline
Nanoparticles could improve cancer diagnosis
Display Headline
Nanoparticles could improve cancer diagnosis
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

Model reveals how to target cancer’s weaknesses

Article Type
Changed
Thu, 07/17/2014 - 05:00
Display Headline
Model reveals how to target cancer’s weaknesses

Drug release in a cancer cell

Credit: PNAS

A new model suggests we should be targeting cancers’ weaknesses instead of their strengths.

An article in BioEssays proposes that cancers form when recently evolved genes are damaged, and cancer cells have to revert to using older, inappropriate genetic pathways.

So we should create treatments that take advantage of capabilities humans have developed more recently—such as the adaptive immune system—instead of trying to target older capabilities—such as the innate immune system and cell proliferation.

“The rapid proliferation of cancer cells is an ancient, default capability that became regulated during the evolution of multicellularity about a billion years ago,” said study author Charley Lineweaver, PhD, of The Australian National University in Canberra.

“Our model suggests that cancer progression is the accumulation of damage to the more recently acquired genes. Without the regulation of these recent genes, cell physiology reverts to earlier programs, such as unregulated cell proliferation.”

To develop their model, Dr Lineweaver and his colleagues turned to knowledge uncovered by genome sequencing in a range of our distant relatives, including fish, coral, and sponges.

This knowledge has allowed scientists to establish the order in which genes evolved and is the basis of the new therapeutic implications of the model, Dr Lineweaver said.

He noted that the standard model of cancer development suggests that selection produces the acquired capabilities of cancer—such as sustained proliferative signaling and evading apoptosis—and they evolve during the lifetime of the patient.

But Dr Lineweaver’s model suggests the capabilities of cancer are acquired atavisms. They are activated during early embryogenesis and wound healing and reactivated inappropriately during carcinogenesis.

The most recent capabilities—mammalian and vertebrate capabilities—are the least entrenched in cancer. So they should be targeted with therapy.

The older capabilities—last eukaryotic common ancestor (LECA) capabilities, stem eukaryote capabilities, and the earliest evolved capabilities—are maintained in cancer and are therefore difficult to target.

For example, some human ATP binding cassette (ABC) transporters are ancient, and some are quite recent. Dr Lineweaver and his colleagues found that older ABC proteins were more likely to be active in cancer.

So the researchers believe we should create treatments that can be expelled by the newer ABC transporters. That way, normal cells will expel the treatment, but cancer cells will not.

Another potential treatment avenue, according to Dr Lineweaver, is targeting the adaptive immune system.

“The adaptive immune system that humans have has evolved relatively recently, and it seems cancer cells do not have the ability to talk to and be protected by it,” he noted.

“The new therapeutic strategies we are proposing target these weaknesses. These strategies are very different from current therapies, which attack cancer’s strength—its ability to proliferate rapidly.”

Publications
Topics

Drug release in a cancer cell

Credit: PNAS

A new model suggests we should be targeting cancers’ weaknesses instead of their strengths.

An article in BioEssays proposes that cancers form when recently evolved genes are damaged, and cancer cells have to revert to using older, inappropriate genetic pathways.

So we should create treatments that take advantage of capabilities humans have developed more recently—such as the adaptive immune system—instead of trying to target older capabilities—such as the innate immune system and cell proliferation.

“The rapid proliferation of cancer cells is an ancient, default capability that became regulated during the evolution of multicellularity about a billion years ago,” said study author Charley Lineweaver, PhD, of The Australian National University in Canberra.

“Our model suggests that cancer progression is the accumulation of damage to the more recently acquired genes. Without the regulation of these recent genes, cell physiology reverts to earlier programs, such as unregulated cell proliferation.”

To develop their model, Dr Lineweaver and his colleagues turned to knowledge uncovered by genome sequencing in a range of our distant relatives, including fish, coral, and sponges.

This knowledge has allowed scientists to establish the order in which genes evolved and is the basis of the new therapeutic implications of the model, Dr Lineweaver said.

He noted that the standard model of cancer development suggests that selection produces the acquired capabilities of cancer—such as sustained proliferative signaling and evading apoptosis—and they evolve during the lifetime of the patient.

But Dr Lineweaver’s model suggests the capabilities of cancer are acquired atavisms. They are activated during early embryogenesis and wound healing and reactivated inappropriately during carcinogenesis.

The most recent capabilities—mammalian and vertebrate capabilities—are the least entrenched in cancer. So they should be targeted with therapy.

The older capabilities—last eukaryotic common ancestor (LECA) capabilities, stem eukaryote capabilities, and the earliest evolved capabilities—are maintained in cancer and are therefore difficult to target.

For example, some human ATP binding cassette (ABC) transporters are ancient, and some are quite recent. Dr Lineweaver and his colleagues found that older ABC proteins were more likely to be active in cancer.

So the researchers believe we should create treatments that can be expelled by the newer ABC transporters. That way, normal cells will expel the treatment, but cancer cells will not.

Another potential treatment avenue, according to Dr Lineweaver, is targeting the adaptive immune system.

“The adaptive immune system that humans have has evolved relatively recently, and it seems cancer cells do not have the ability to talk to and be protected by it,” he noted.

“The new therapeutic strategies we are proposing target these weaknesses. These strategies are very different from current therapies, which attack cancer’s strength—its ability to proliferate rapidly.”

Drug release in a cancer cell

Credit: PNAS

A new model suggests we should be targeting cancers’ weaknesses instead of their strengths.

An article in BioEssays proposes that cancers form when recently evolved genes are damaged, and cancer cells have to revert to using older, inappropriate genetic pathways.

So we should create treatments that take advantage of capabilities humans have developed more recently—such as the adaptive immune system—instead of trying to target older capabilities—such as the innate immune system and cell proliferation.

“The rapid proliferation of cancer cells is an ancient, default capability that became regulated during the evolution of multicellularity about a billion years ago,” said study author Charley Lineweaver, PhD, of The Australian National University in Canberra.

“Our model suggests that cancer progression is the accumulation of damage to the more recently acquired genes. Without the regulation of these recent genes, cell physiology reverts to earlier programs, such as unregulated cell proliferation.”

To develop their model, Dr Lineweaver and his colleagues turned to knowledge uncovered by genome sequencing in a range of our distant relatives, including fish, coral, and sponges.

This knowledge has allowed scientists to establish the order in which genes evolved and is the basis of the new therapeutic implications of the model, Dr Lineweaver said.

He noted that the standard model of cancer development suggests that selection produces the acquired capabilities of cancer—such as sustained proliferative signaling and evading apoptosis—and they evolve during the lifetime of the patient.

But Dr Lineweaver’s model suggests the capabilities of cancer are acquired atavisms. They are activated during early embryogenesis and wound healing and reactivated inappropriately during carcinogenesis.

The most recent capabilities—mammalian and vertebrate capabilities—are the least entrenched in cancer. So they should be targeted with therapy.

The older capabilities—last eukaryotic common ancestor (LECA) capabilities, stem eukaryote capabilities, and the earliest evolved capabilities—are maintained in cancer and are therefore difficult to target.

For example, some human ATP binding cassette (ABC) transporters are ancient, and some are quite recent. Dr Lineweaver and his colleagues found that older ABC proteins were more likely to be active in cancer.

So the researchers believe we should create treatments that can be expelled by the newer ABC transporters. That way, normal cells will expel the treatment, but cancer cells will not.

Another potential treatment avenue, according to Dr Lineweaver, is targeting the adaptive immune system.

“The adaptive immune system that humans have has evolved relatively recently, and it seems cancer cells do not have the ability to talk to and be protected by it,” he noted.

“The new therapeutic strategies we are proposing target these weaknesses. These strategies are very different from current therapies, which attack cancer’s strength—its ability to proliferate rapidly.”

Publications
Publications
Topics
Article Type
Display Headline
Model reveals how to target cancer’s weaknesses
Display Headline
Model reveals how to target cancer’s weaknesses
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

New insight into MYC-induced lymphoma

Article Type
Changed
Wed, 07/16/2014 - 06:00
Display Headline
New insight into MYC-induced lymphoma

MYC-expressing cancer cells

Credit: Juha Klefstrom

Investigators have identified biological signatures in lymphoma cells that can be traced back to the original oncogene.

The team analyzed mouse models and patient samples of MYC-induced lymphoma. And they discovered lipid signatures that corresponded with the level of MYC expression.

The investigators believe this discovery could be the first step toward developing a technique to identify the origin of lymphomas and other malignancies.

They described their discovery in PNAS.

“The same cancer can occur because of different genes, but, in certain cases, the aggressiveness and the type of treatment actually depend a lot on what oncogene caused that cancer,” said study author Livia Eberlin, PhD, of Stanford University in California.

With that in mind, she and her colleagues looked at MYC, an oncogene that’s responsible for approximately half of all human cancers. They wanted to find a biological signature that would trace the mutating cancer cells back to the original oncogene.

“When cancer takes place, the cell loves to gobble up glucose—that’s a sugar—and glutamine,” said Richard Zare, PhD, also of Stanford. “It takes those and makes different lipids—different fatty molecules than what it normally makes.”

So the investigators set out to evaluate changes in lipid profiles in MYC-induced lymphoma. They compared lipid signatures in MYC-induced transgenic mouse models to those in normal control mice.

The team identified 104 molecular ions that were either increased or decreased in the MYC lymphoma models compared to controls. And 86 of these ions were complex phospholipids.

Most of the lipids that were increased in lymphoma were glycerophosphoglycerols and cardiolipins, with a higher content of monounsaturated fatty acids when compared with controls.

To determine if these findings might also apply to humans, the investigators examined 15 samples from lymphoma patients.

The samples had varying expression levels of MYC oncoprotein, and the team observed distinct lipid profiles in lymphomas with high and low MYC expression. This included many of the lipid species they had identified in the animal models of MYC-induced lymphoma.

The investigators said their results suggest a relationship between specific lipid species and the overexpression of MYC. And this information could have both diagnostic and prognostic applications.

Publications
Topics

MYC-expressing cancer cells

Credit: Juha Klefstrom

Investigators have identified biological signatures in lymphoma cells that can be traced back to the original oncogene.

The team analyzed mouse models and patient samples of MYC-induced lymphoma. And they discovered lipid signatures that corresponded with the level of MYC expression.

The investigators believe this discovery could be the first step toward developing a technique to identify the origin of lymphomas and other malignancies.

They described their discovery in PNAS.

“The same cancer can occur because of different genes, but, in certain cases, the aggressiveness and the type of treatment actually depend a lot on what oncogene caused that cancer,” said study author Livia Eberlin, PhD, of Stanford University in California.

With that in mind, she and her colleagues looked at MYC, an oncogene that’s responsible for approximately half of all human cancers. They wanted to find a biological signature that would trace the mutating cancer cells back to the original oncogene.

“When cancer takes place, the cell loves to gobble up glucose—that’s a sugar—and glutamine,” said Richard Zare, PhD, also of Stanford. “It takes those and makes different lipids—different fatty molecules than what it normally makes.”

So the investigators set out to evaluate changes in lipid profiles in MYC-induced lymphoma. They compared lipid signatures in MYC-induced transgenic mouse models to those in normal control mice.

The team identified 104 molecular ions that were either increased or decreased in the MYC lymphoma models compared to controls. And 86 of these ions were complex phospholipids.

Most of the lipids that were increased in lymphoma were glycerophosphoglycerols and cardiolipins, with a higher content of monounsaturated fatty acids when compared with controls.

To determine if these findings might also apply to humans, the investigators examined 15 samples from lymphoma patients.

The samples had varying expression levels of MYC oncoprotein, and the team observed distinct lipid profiles in lymphomas with high and low MYC expression. This included many of the lipid species they had identified in the animal models of MYC-induced lymphoma.

The investigators said their results suggest a relationship between specific lipid species and the overexpression of MYC. And this information could have both diagnostic and prognostic applications.

MYC-expressing cancer cells

Credit: Juha Klefstrom

Investigators have identified biological signatures in lymphoma cells that can be traced back to the original oncogene.

The team analyzed mouse models and patient samples of MYC-induced lymphoma. And they discovered lipid signatures that corresponded with the level of MYC expression.

The investigators believe this discovery could be the first step toward developing a technique to identify the origin of lymphomas and other malignancies.

They described their discovery in PNAS.

“The same cancer can occur because of different genes, but, in certain cases, the aggressiveness and the type of treatment actually depend a lot on what oncogene caused that cancer,” said study author Livia Eberlin, PhD, of Stanford University in California.

With that in mind, she and her colleagues looked at MYC, an oncogene that’s responsible for approximately half of all human cancers. They wanted to find a biological signature that would trace the mutating cancer cells back to the original oncogene.

“When cancer takes place, the cell loves to gobble up glucose—that’s a sugar—and glutamine,” said Richard Zare, PhD, also of Stanford. “It takes those and makes different lipids—different fatty molecules than what it normally makes.”

So the investigators set out to evaluate changes in lipid profiles in MYC-induced lymphoma. They compared lipid signatures in MYC-induced transgenic mouse models to those in normal control mice.

The team identified 104 molecular ions that were either increased or decreased in the MYC lymphoma models compared to controls. And 86 of these ions were complex phospholipids.

Most of the lipids that were increased in lymphoma were glycerophosphoglycerols and cardiolipins, with a higher content of monounsaturated fatty acids when compared with controls.

To determine if these findings might also apply to humans, the investigators examined 15 samples from lymphoma patients.

The samples had varying expression levels of MYC oncoprotein, and the team observed distinct lipid profiles in lymphomas with high and low MYC expression. This included many of the lipid species they had identified in the animal models of MYC-induced lymphoma.

The investigators said their results suggest a relationship between specific lipid species and the overexpression of MYC. And this information could have both diagnostic and prognostic applications.

Publications
Publications
Topics
Article Type
Display Headline
New insight into MYC-induced lymphoma
Display Headline
New insight into MYC-induced lymphoma
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica