Pfizer Presents Updated Favorable Elranatamab Data from Pivotal Phase 2 MagnetisMM-3 Trial

On December 10, 2022 Pfizer Inc. (NYSE:PFE) reported 10.4 month follow-up data from the pivotal Phase 2 MagnetisMM-3 clinical trial suggesting elranatamab, a B-cell maturation antigen (BCMA)-CD3-targeted bispecific antibody (BsAb), is efficacious and has a manageable safety profile in patients with relapsed or refractory multiple myeloma (RRMM) in a heavily pretreated population, who have received at least three classes of prior therapies including a proteasome inhibitor, an immunomodulatory agent, and an anti-CD38 monoclonal antibody (i.e. triple-class refractory or exposed) (Press release, Pfizer, DEC 10, 2022, View Source [SID1234625059]). These data are being presented today in an oral session at the 64th American Society of Hematology (ASH) (Free ASH Whitepaper) Annual Meeting and Exposition 2022 (abstract 159).

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

"Forward-Looking Information and Factors That May Affect Future Results"

Tweet this
"Although there are approved treatments for multiple myeloma, none are currently curative, and patients often find themselves with dwindling therapeutic options as their disease relapses or becomes refractory to successive medicines," said Nizar Bahlis, M.D., Associate Professor, Arnie Charbonneau Cancer Institute, University of Calgary, Canada, and lead investigator. "These longer-term results show early and deep responses with elranatamab in a heavily pretreated patient population, adding to the growing body of evidence showing elranatamab has the potential to be a transformative option in an area of high need."

Elranatamab is an investigational humanized BsAb that targets both BCMA-expressing multiple myeloma cells and CD3-expressing T-cells, bridging them together and activating the T-cells to kill the myeloma cells. The binding affinities of elranatamab for BCMA and CD3 have been engineered to elicit potent T-cell-mediated anti-myeloma activity. Given factors currently limiting the availability of novel therapies in the triple-class exposed setting, elranatamab has the potential to reach a broader and greater number of patients as an off-the-shelf option that is administered subcutaneously (SC), which offers more convenience over intravenous administration.

In this analysis, safety and efficacy were analyzed in 123 patients who had received at least one dose of elranatamab (cohort A – BCMA-naïve) as of the data cut-off on October 14, 2022. Patients received SC elranatamab 76 mg weekly (QW) on a 28-day cycle with a step-up priming dose regimen, 12 mg and 32 mg administered on Day 1 and Day 4, respectively, during Cycle 1. With a median follow up of 10.4 months, patients who received elranatamab achieved a high objective response rate of 61%, with 84% probability of maintaining response at nine months. Probability of progression-free survival and overall survival were 63% and 70%, respectively, at nine months. The results also suggest elranatamab has a manageable safety profile and that the two-step-up priming dose regimen (12/32 mg) mitigated the rate and severity of cytokine release syndrome (CRS) (58%, all Grade 1/2) and immune effector cell-associated neurotoxicity syndrome (ICANS, 3%, all Grade 1/2) in cohort A of MagnetisMM-3 (n=119). The most common hematologic treatment emergent adverse events (TEAEs) of any grade were – anemia (48%), neutropenia (48%), thrombocytopenia (30%) and lymphopenia (26%).

"Discovered and developed at Pfizer, elranatamab is just one example of our focus on investing in breakthrough science. We’re applying our expertise in hematology developed over a decade to advance elranatamab as an innovative treatment for multiple myeloma," said Chris Boshoff, M.D., Ph.D., Chief Development Officer, Oncology and Rare Disease, Pfizer Global Product Development. "We are excited to be sharing our latest research at ASH (Free ASH Whitepaper), which further supports the efficacy and safety of elranatamab, and will continue to evaluate its potential benefits through the MagnetisMM clinical trial program across earlier and broader populations, including newly diagnosed patients. We are excited by the potential for elranatamab, if approved, to reach a greater number of patients globally across the treatment paradigm."

Additional Pfizer elranatamab presentations at ASH (Free ASH Whitepaper) include:

Elranatamab, a BCMA Targeted T-Cell Engaging Bispecific Antibody, Induces Durable Clinical and Molecular Responses for Patients with RRMM (abstract 158, oral)
MagnetisMM-1 Phase 1/2 first in human MagnetisMM-1 study (NCT03269136)
Safety and preliminary efficacy results from Part 1 (safety lead-in cohort) of MagnetisMM-5, a Phase 3 study of elranatamab + daratumumab in patients with RRMM (NCT05020236) (poster 1921)
MagnetisMM-4: An Open Label, Phase 1b/2 Umbrella Study of Elranatamab in Combination with Other Anti-cancer Treatments for Patients with Multiple Myeloma (poster 4567)
Phase 1b/2 MagnetisMM-4 (NCT05090566) ongoing clinical trial
Dose Optimization to Mitigate the Risk of CRS with Elranatamab in Multiple Myeloma (poster 3192)
Dose-optimization analysis from across MagnetisMM-1 (NCT03269136), MagnetisMM-2 (NCT04798586), MagnetisMM-3 (NCT04649359) and MagnetisMM-9 (NCT05014412)
A Systematic Meta-analysis of Cytokine Release Syndrome Incidence in B-Cell Maturation Antigen-Targeting Chimeric Antigen Receptor T-Cell Therapy and Bispecific Antibodies for Patients with Relapsed and/or Refractory Multiple Myeloma (poster 4509)
Meta-analysis of CRS among patients treated with BCMA-targeting CAR-Ts and BsAbs across 53 studies
MagnetisMM-3 (NCT04649359) is an ongoing open-label, multicenter, non-randomized study designed to evaluate the safety and efficacy of elranatamab as monotherapy in patients with RRMM. Additional data continue to be collected and will be shared as they mature. Prior to enrollment, participants had received a median of five lines of treatment. The participants included in this study represent a difficult-to-treat multiple myeloma patient population; almost all (97%) of the 123 patients included in this analysis were triple-class refractory and nearly half (42%) were penta-drug refractory*. The trial is part of the robust MagnetisMM multiple indication clinical research program that expands to additional patient populations over time, with ongoing registration-intent trials that explore elranatamab both as monotherapy and in combination with standard or novel therapies, spanning multiple patient populations from newly diagnosed multiple myeloma, double-class exposed disease and RRMM.

In November 2022, Pfizer announced elranatamab received Breakthrough Therapy Designation from the Food and Drug Administration (FDA) for the treatment of RRMM. Elranatamab has also been granted Orphan Drug Designation by the FDA and the European Medicines Agency (EMA) for the treatment of MM. The FDA and EMA have granted elranatamab Fast Track Designation and the PRIME scheme, respectively, for the treatment of patients with RRMM. The UK Medicines and Healthcare Products Regulatory Agency (MHRA) has also granted elranatamab Innovative Medicine Designation and the Innovation Passport for the treatment of MM.

* Penta-drug refers to at least 2 proteosome inhibitors, 2 immunomodulatory drugs, and 1 anti-CD38 antibody.

About Multiple Myeloma

Multiple myeloma is a blood cancer that affects plasma cells made in the bone marrow. Healthy plasma cells make antibodies that help the body fight infection. There are over 34,000 new cases of multiple myeloma diagnosed annually in the U.S. and 176,000 globally.1,2 Despite treatment advances, there is currently no cure. The median overall survival is just over five years, and most patients receive four or more lines of therapy.3

Orca Bio Presents Data Demonstrating its Lead Investigational High-Precision Cell Therapy Orca-T Significantly Improved Relapse-Free Survival at 64th ASH Annual Meeting

On December 10, 2022 Orca Bio, a late-stage biotechnology company developing high-precision cell therapies for the treatment of cancer, genetic blood disorders and autoimmune diseases, reported updated positive clinical data on its lead investigational cell therapy, Orca-T, during an oral presentation at the 64th American Society of Hematology (ASH) (Free ASH Whitepaper) Annual Meeting (Press release, Orca Bio, DEC 10, 2022, View Source [SID1234625058]).

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

The data demonstrated a significant improvement in relapse-free survival (RFS) at one year with 87% in 71 patients who received Orca-T and busulfan, fludarabine and thiotepa (BFT) conditioning. BFT is the preferred conditioning regimen used in Precision-T, the ongoing Phase 3 pivotal study of Orca-T. Additionally, across all 151 patients, there was an increase in graft-versus-host disease-free, relapse-free survival (GRFS) rates and overall survival rates at one year with Orca-T compared to an independent cohort.

"Today, providers must balance treatment decisions with the goal of fighting cancer while minimizing toxicities. With a standard allogeneic transplant, highly myeloablative conditioning regimens can lead to reduced rates of patient relapse, but can also increase rates of non-relapse mortality," said Everett Meyer, M.D., Ph.D., primary investigator. "It is encouraging to see that the use of Orca-T across multiple hematological malignancies resulted in a remarkable improvement in relapse-free and overall survival while lowering the rate of serious toxicities compared to standard of care allogeneic hematopoietic stem cell transplants."

The results of the patient subgroup conditioned with BFT were presented today along with pooled results from the single-center Phase 2 and multi-center Phase 1b trials from 151 patients treated with Orca-T with acute myeloid leukemia (AML), acute lymphocytic leukemia (ALL), myelodysplastic syndromes (MDS) and other hematological malignancies. For comparison purposes, an independent Center for International Blood and Marrow Transplant Research (CIBMTR)-based cohort was identified consisting of patients with AML, ALL or MDS who received a standard of care allogeneic hematopoietic stem cell transplant.

Results demonstrated that patients conditioned with BFT and treated with Orca-T experienced:

87% RFS at one year among all groups, including minimal residual disease positive acute leukemia patients.
81% GRFS at one year.
0% non-relapse mortality and only 5% moderate-to-severe chronic graft versus host disease at one year.
94% overall survival (OS) at one year.
In pooled results from all patients treated with Orca-T, patients experienced:

70% GRFS at one year compared to 21% in the CIBMTR-based cohort.
4% non-relapse mortality at one year compared to 10% in the CIBMTR-based cohort.
88% OS at one year compared to 68% in the CIBMTR-based cohort.
Importantly, Orca-T was manufactured reliably and delivered with vein-to-vein times of 72 hours or less across the U.S.

"These relapse-free and overall survival rates from more than 150 patients treated with Orca-T are very encouraging, and we are extremely pleased to see the strong results of patients who were given the preferred conditioning regimen used in our pivotal Phase 3 trial," said Ivan Dimov, Ph.D., co-founder and chief executive officer of Orca Bio. "These new data continue to reinforce the potential of this high-precision cell therapy to become a better treatment option for patients and transplant physicians. We remain laser-focused on advancing Orca-T through Precision-T and ultimately on delivering the therapy to more patients in need."

The full presentation will be made available on www.orcabio.com.

About Orca-T
Orca-T is an investigational high-precision allogeneic cellular therapy consisting of infusions containing regulatory T-cells, conventional T-cells and CD34+ stem cells derived from peripheral blood from either related or unrelated matched donors. Orca-T has received Regenerative Medicine Advanced Therapy (RMAT) designation from the U.S. Food and Drug Administration and is being studied to treat multiple hematologic malignancies.

Incyte Announces Data from Two LIMBER Studies Evaluating Combination Treatments in Patients with Myelofibrosis (MF) Presented at ASH 2022

On December 10, 2022 Incyte (Nasdaq:INCY) reported new data from two of its LIMBER (Leadership In MPNs and GVHD BEyond Ruxolitinib) trials evaluating monotherapy and combination strategies using ruxolitinib (Jakafi) with parsaclisib, its investigational phosphatidylinositol 3-kinase delta (PI3Kδ) inhibitor, and INCB00928 (zilurgisertib), its activin receptor-like kinase (ALK2) inhibitor, in patients with myelofibrosis (MF) (Press release, Incyte, DEC 10, 2022, View Source [SID1234625057]). These Phase 2 and Phase 1/2 trials (Abstract #236 and Abstract #1714, respectively) were presented at the 64th American Society of Hematology (ASH) (Free ASH Whitepaper) Annual Meeting, held December 10-13, 2022, in New Orleans and virtually1,2.

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

"MF is a rare, chronic blood cancer, and despite the advancements made in treatment, additional options are needed"

"Despite the significant advances we have made in the treatment of myeloproliferative neoplasms (MPNs) like MF, a need for additional options remains for those who have an inadequate response to or are unable to tolerate current therapies," said Peter Langmuir, M.D., Vice President, Oncology Drug Development, Incyte. "The parsaclisib and ruxolitinib data presented at ASH (Free ASH Whitepaper) demonstrate the clinical potential of the combination to improve upon the standard of care and continue to support the safety profile. We look forward to building on these results through our Phase 3 LIMBER-304 and LIMBER-313 trials evaluating parsaclisib as an add-on to ruxolitinib and in the frontline setting, both of which are currently underway."

Final results from the Phase 2 trial (Abstract #236; NCT02718300) evaluating the efficacy and safety of add-on parsaclisib to ruxolitinib for patients with MF who had a suboptimal response to ruxolitinib resulted in additional spleen volume reduction and improvement in symptom burden with add-on parsaclisib. Patients in the trial received parsaclisib daily for eight weeks in combination with stable dose ruxolitinib and then daily or weekly thereafter. Patients who received an all daily parsaclisib dosing schedule appeared to have a more durable efficacy profile compared with daily followed by weekly dosing of parsaclisib. Specifically:

At 12 weeks of treatment, 59.5% (25), 21.4% (9) and 4.8% (2) of patients who received all daily dosing experienced ≥10%, ≥25% and ≥35% reduction in spleen volume, respectively.
Comparatively, 28.1% (9), 3.1% (1) and 0% of patients who received daily followed by weekly dosing experienced ≥10%, ≥25% and ≥35% reduction in spleen volume, respectively.
At 24 weeks of treatment the reduction was maintained, with 50% (21), 28.6% (12) and 7.1% (3) patients who received all daily dosing experiencing ≥10%, ≥25% and ≥35% reduction in spleen volume, respectively.
Comparatively, 12.5% (4), 12.5% (4) and 3.1% (1) of patients who received daily followed by weekly dosing experienced ≥10%, ≥25% and ≥35% reduction in spleen volume, respectively.
Addition of parsaclisib to ruxolitinib was generally well-tolerated, with limited grade 3 or 4 adverse events and treatment-emergent adverse event (TEAE)-related discontinuations. TEAEs common to PI3Kδ inhibitors in lymphoma (e.g., hepatotoxicity, rash, colitis) were infrequent or absent with the addition of parsaclisib.
Serious TEAEs occurring in ≥2 patients overall included pneumonia (n=6; 2 daily/weekly, 1 all daily), fall (n=3; 2 daily/weekly, 1 all daily) and pyrexia (n=2; 1 daily/weekly, 1 all daily).
Overall, 9 patients (5 daily/weekly, 4 all daily) had a TEAE leading to parsaclisib discontinuation, and 4 patients (2 daily/weekly, 2 all daily) had a TEAE leading to ruxolitinib discontinuation.
"MF is a rare, chronic blood cancer, and despite the advancements made in treatment, additional options are needed," said Abdulraheem Yacoub, Associate Professor, Hematologic Malignancies and Cellular Therapeutics, University of Kansas Cancer Center. "I am encouraged by these findings and the potential of parsaclisib and ruxolitinib to be an efficacious combination therapy to help improve outcomes for certain patients living with MF."

Additionally, data from a Phase 1/2 open-label, dose escalation and expansion study (Abstract #1714; NCT04455841) assessing the safety and tolerability of INCB00928 (zilurgisertib), a potent and selective ALK2 inhibitor, as monotherapy or in combination with ruxolitinib in patients with anemia due to MF were presented at ASH (Free ASH Whitepaper). Anemia occurs in more than one-third of patients at MF diagnosis and can be exacerbated during treatment3,4. Initial results of the study observed reduction in post-dose hepcidin levels at all dose levels and observed improvements in anemia among patients treated in both the monotherapy and combination cohorts, which suggest the potential for therapeutic activity. The data also support once-daily dosing of INCB00928 and continued dose escalation to achieve optimal exposure. Treatment with INCB00928 monotherapy and in combination with ruxolitinib resulted in predominantly grade 1/2 TEAEs and no dose-limiting toxicities (DLTs). Few grade ≥3 TEAEs were observed, including thrombocytopenia in two patients with baseline grade 2 thrombocytopenia, and neutropenia in one patient with baseline grade 2 neutropenia. No TEAEs led to study drug discontinuation.

More information regarding the congress and the more than 50 abstracts from Incyte’s oncology portfolio being featured at the meeting is available on the ASH (Free ASH Whitepaper) website: View Source

About Myeloproliferative Neoplasms

Myeloproliferative neoplasms (MPNs) are a closely related group of blood cancers in which the bone marrow functions abnormally. The bone marrow is where the body’s blood cells are made. MPNs are progressive blood cancers that can strike anyone at any age, but they are more common in older adults. Estimates of the prevalence of MPNs vary, but analysis of claims data suggests there may be as many as 200,000 people in the U.S. living with the most prevalent MPNs: myelofibrosis, polycythemia vera or essential thrombocythemia5.

About LIMBER

Incyte is a leader in the discovery and development of therapies for patients with myeloproliferative neoplasms (MPNs) and graft-versus-host disease (GVHD). The Leadership In MPNs and GVHD BEyond Ruxolitinib (LIMBER) program is designed to evaluate multiple monotherapy and combination strategies to improve and expand treatments for patients with MPNs and GVHD. The program currently has three key areas of focus: development of a new, once-daily formulation of ruxolitinib; ruxolitinib-based combinations with new targets such as PI3Kδ, BET and ALK2; and new therapeutic options such as mutant CALR.

About Jakafi (ruxolitinib)

Jakafi (ruxolitinib) is a JAK1/JAK2 inhibitor approved by the U.S. FDA for treatment of polycythemia vera (PV) in adults who have had an inadequate response to or are intolerant of hydroxyurea; intermediate or high-risk myelofibrosis (MF), including primary MF, post-polycythemia vera MF and post-essential thrombocythemia MF in adults; steroid-refractory acute GVHD in adult and pediatric patients 12 years and older; and chronic GVHD after failure of one or two lines of systemic therapy in adult and pediatric patients 12 years and older6.

Jakafi is marketed by Incyte in the United States and by Novartis as Jakavi (ruxolitinib) outside the United States. Jakafi is a registered trademark of Incyte Corporation. Jakavi is a registered trademark of Novartis AG in countries outside the United States.

Important Safety Information

Jakafi can cause serious side effects, including:

Low blood counts: Jakafi (ruxolitinib) may cause low platelet, red blood cell, and white blood cell counts. If you develop bleeding, stop taking Jakafi and call your healthcare provider. Your healthcare provider will do a blood test to check your blood counts before you start Jakafi and regularly during your treatment. Your healthcare provider may change your dose of Jakafi or stop your treatment based on the results of your blood tests. Tell your healthcare provider right away if you develop or have worsening symptoms such as unusual bleeding, bruising, tiredness, shortness of breath, or a fever.

Infection: You may be at risk for developing a serious infection during treatment with Jakafi. Tell your healthcare provider if you develop any of the following symptoms of infection: chills, nausea, vomiting, aches, weakness, fever, painful skin rash or blisters.

Cancer: Some people have had certain types of non-melanoma skin cancers during treatment with Jakafi. Your healthcare provider will regularly check your skin during your treatment with Jakafi. Tell your healthcare provider if you develop any new or changing skin lesions during treatment with Jakafi.

Increases in cholesterol: You may have changes in your blood cholesterol levels during treatment with Jakafi. Your healthcare provider will do blood tests to check your cholesterol levels about every 8 to 12 weeks after you start taking Jakafi, and as needed.

Increased risk of major cardiovascular events such as heart attack, stroke or death in people who have cardiovascular risk factors and who are current or past smokers while using another JAK inhibitor to treat rheumatoid arthritis: Get emergency help right away if you have any symptoms of a heart attack or stroke while taking Jakafi, including: discomfort in the center of your chest that lasts for more than a few minutes, or that goes away and comes back, severe tightness, pain, pressure, or heaviness in your chest, throat, neck, or jaw, pain or discomfort in your arms, back, neck, jaw, or stomach, shortness of breath with or without chest discomfort, breaking out in a cold sweat, nausea or vomiting, feeling lightheaded, weakness in one part or on one side of your body, slurred speech

Increased risk of blood clots: Blood clots in the veins of your legs (deep vein thrombosis, DVT) or lungs (pulmonary embolism, PE) have happened in people taking another JAK inhibitor for rheumatoid arthritis and may be life-threatening. Tell your healthcare provider right away if you have any signs and symptoms of blood clots during treatment with Jakafi, including: swelling, pain, or tenderness in one or both legs, sudden, unexplained chest or upper back pain, shortness of breath or difficulty breathing

Possible increased risk of new (secondary) cancers: People who take another JAK inhibitor for rheumatoid arthritis have an increased risk of new (secondary) cancers, including lymphoma and other cancers. People who smoke or who smoked in the past have an added risk of new cancers.

The most common side effects of Jakafi include: for certain types of myelofibrosis (MF) and polycythemia vera (PV) – low platelet or red blood cell counts, bruising, dizziness, headache, and diarrhea; for acute GVHD – low platelet counts, low red or white blood cell counts, infections, and swelling; and for chronic GVHD – low red blood cell or platelet counts and infections including viral infections.

These are not all the possible side effects of Jakafi. Ask your pharmacist or healthcare provider for more information. Call your doctor for medical advice about side effects.

Before taking Jakafi, tell your healthcare provider about: all the medications, vitamins, and herbal supplements you are taking and all your medical conditions, including if you have an infection, have or had low white or red blood cell counts, have or had tuberculosis (TB) or have been in close contact with someone who has TB, had shingles (herpes zoster), have or had hepatitis B, have or had liver or kidney problems, are on dialysis, have high cholesterol or triglycerides, had cancer, are a current or past smoker, had a blood clot, heart attack, other heart problems or stroke, or have any other medical condition. Take Jakafi exactly as your healthcare provider tells you. Do not change your dose or stop taking Jakafi without first talking to your healthcare provider.

Women should not take Jakafi while pregnant or planning to become pregnant. Do not breastfeed during treatment with Jakafi and for 2 weeks after the final dose.

Please see the Full Prescribing Information, which includes a more complete discussion of the risks associated with Jakafi.

You are encouraged to report negative side effects of prescription drugs to the FDA. Visit www.fda.gov/medwatch, or call 1-800-FDA-1088.

You may also report side effects to Incyte Medical Information at 1-855-463-3463.

New Results from the Phase 3 GLOW Study of Fixed-Duration Treatment with IMBRUVICA® (ibrutinib) Plus Venetoclax Demonstrate Robust Efficacy and Sustained Response in Older, Unfit Patients with Previously Untreated Chronic Lymphocytic Leukemia

On December 10, 2022 The Janssen Pharmaceutical Companies of Johnson & Johnson reported new four-year follow-up results from the Phase 3 GLOW study (Abstract #93), which showed investigational, fixed-duration treatment with IMBRUVICA + venetoclax (I+V) reduced the risk of progression or death by 79 percent among older and/or unfit patients with previously untreated chronic lymphocytic leukemia (CLL) compared to patients treated with chemoimmunotherapy (Press release, Janssen Biotech, DEC 10, 2022, View Source [SID1234625042]).1 These results were highlighted in an oral presentation during the 2022 American Society of Hematology (ASH) (Free ASH Whitepaper) Annual Meeting.1

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

CLL is the most common form of leukemia in adults in the U.S. and currently has no cure.2 While the treatment landscape has evolved significantly since the emergence of targeted agents, there is still significant unmet need for novel treatment options, including fixed-duration regimens.

"The GLOW study results demonstrate the potential of fixed-duration I+V to become an additional treatment option for older, unfit patients with CLL in the first-line setting, and this fixed-dose combination may offer a flexible regimen for patients seeking a time-limited treatment approach," said study investigator Carsten Niemann,† M.D., Ph.D., Clinical Associate Professor and Principal Investigator at Rigshospitalet, Copenhagen, Denmark. "This first all-oral, fixed-duration novel combination demonstrates an OS advantage in the first-line treatment of CLL and is an innovative option for patients."

In the study, fixed-duration I+V therapy exhibited robust efficacy in older and/or unfit adults with previously untreated CLL, with a superior and sustained benefit in progression-free survival (PFS) with four years of follow-up.1 Seventy-five percent of patients treated with the combination were alive and progression-free at 3.5 years.1 I+V also demonstrated an OS advantage versus chlorambucil plus obinutuzumab (Clb+O) at this latest study follow-up.1 Exploratory analyses showed that post-treatment PFS rates were higher for I+V (n=106) than Clb+O (n=105), regardless of minimal residual disease (MRD) status post-treatment.1

GLOW Results

With a median 46 months of follow-up, I+V reduced the risk of disease progression or death by 79 percent versus Clb+O (Hazard Ratio (HR) 0.214; [95 percent Confidence Interval (CI), 0.138-0.334]; p<0.0001).1
I+V is the first fixed-duration novel combination to demonstrate an OS advantage compared to Clb+O in the first-line treatment of CLL (HR 0.487; [95 percent CI, 0.262-0.907]; nominal p=0.0205).1
An estimated 74.6 percent of previously untreated older and/or comorbid patients were alive and progression-free at 3.5 years with all-oral, once-daily, fixed-duration I+V treatment compared to an estimated 24.8 percent of patients in the Clb+O cohort.1
PFS at 3.5 years was higher for patients in the I+V arm compared to the Clb+O arm for both unmutated IGHV (uIGHV) and mutated IGHV (mIGHV) CLL.1
PFS was better sustained in the I+V arm compared to the Clb+O arm, regardless of MRD (≥10-4) status, measured at three months following end of treatment.1
Two years after end of treatment, estimated PFS was ≥ 90 percent for patients with mIGHV CLL, independent of MRD status, and for the 60 percent of patients with uIGHV CLL who achieved undetectable MRD (uMRD).1
Updated data for both studies showed the safety profile of the I+V regimen was consistent with known safety profiles of IMBRUVICA and venetoclax.1

"IMBRUVICA has helped change the standard of care for adults living with CLL and other B-cell malignancies, and this study adds to the extensive body of evidence supporting its potential to provide improved survival for patients with CLL. These results highlight the potential for front-line CLL patients to be treated with an all-oral, fixed-duration treatment," said Craig Tendler, M.D., Vice President, Late Development and Global Medical Affairs, Janssen Research & Development, LLC. "At Janssen, we are committed to addressing the unmet needs of patients with CLL through continued investment in our IMBRUVICA clinical development program."

In addition to the GLOW results, data from the Phase 2 CAPTIVATE study of the MRD cohort (PCYC-1142) (Abstract #92), which utilized the same I+V schedule for the first 15 cycles as in the GLOW study, were also presented in an oral session at ASH (Free ASH Whitepaper).3 After the first 15 cycles, patients with confirmed uMRD (n=86) were randomized to IMBRUVICA or placebo.3 Disease-free survival at three years post-randomization was 93 percent for IMBRUVICA and 85 percent for placebo. PFS rates at four years from start of treatment were 95 percent and 88 percent, respectively, and OS rates were 98 percent and 100 percent.3

During the three-year post-randomization period, no new atrial fibrillation events occurred in the placebo arm, and no new grade three or higher hemorrhage events occurred in either arm.3 The incidences of hypertension, arthralgia, neutropenia and diarrhea were generally infrequent in the placebo arm during this time period.3 No deaths occurred in either arm during the last 12 months of follow-up.3

About GLOW
The GLOW study (NCT03462719) is a randomized, open-label, Phase 3 trial that evaluated the efficacy and safety of first-line, fixed-duration I+V versus Clb+O in elderly patients (≥65 years of age) with CLL/SLL, or patients ages 18-64 with a cumulative illness rating scale (CIRS) score of greater than six or creatinine clearance less than 70 mL/min, who had active disease requiring treatment per the International Workshop on CLL (iwCLL) criteria.4 Patients with del(17p) or known TP53 mutations were excluded.4 There were 211 patients randomly assigned in a 1:1 ratio to receive either I+V (n=106) and or Clb+O (n=105) and the median age was 71 years.4 Patients assigned to I+V received treatment for 15 cycles (1 cycle is 28 days), starting with three cycles of IMBRUVICA monotherapy lead-in followed by the combination of I+V for 12 cycles.4 Patients assigned to Clb+O were treated for six cycles.4

Among patients with partial response or better, MRD in peripheral blood (PB) was evaluated using next-generation sequencing (NGS) via clonoSEQ on-treatment and at 3-6 month intervals post-treatment.4 ClonoSEQ data was used as part of CLL clonal testing.4

The primary endpoint was PFS up to two years and 10 months.4 Secondary endpoints of the study include MRD negative rate, compete response rate, overall response rate, OS, duration of response and time-to-next treatment.4

About CAPTIVATE

The Phase 2 CAPTIVATE study (NCT02910583) evaluated previously untreated adult patients with CLL who were 70 years or younger, including patients with high-risk disease, in two cohorts: an MRD-guided cohort (n=164; median age, 58 years) and a fixed-duration cohort (n=159; median age, 60 years). Patients received three cycles of IMBRUVICA lead-in followed by 12 cycles of I+V (oral IMBRUVICA [420 mg/d]; oral venetoclax [five-week ramp-up to 400 mg/d]) and the primary endpoint was one-year disease-free survival.5 In this MRD cohort, after completion of I+V, patients with confirmed uMRD were randomly assigned to double-blind treatment with placebo (i.e., a fixed-duration regimen), or continuous IMBRUVICA.5

About IMBRUVICA
IMB­RUVICA (ibrutinib) is a once-daily oral medication that is jointly developed and commercialized by Janssen Biotech, Inc. and Pharmacyclics LLC, an AbbVie company. IMBRUVICA blocks the Bruton’s tyrosine kinase (BTK) protein, which is needed by normal and abnormal B cells, including specific cancer cells, to multiply and spread. By blocking BTK, IMBRUVICA may help move abnormal B cells out of their nourishing environments and inhibits their proliferation.6,7,8

IMBRUVICA is approved in more than 100 countries and has been used to treat more than 270,000 patients worldwide. There are more than 50 company-sponsored clinical trials, including 18 Phase 3 studies, more than 11 years evaluating the efficacy and safety of IMBRUVICA.

IMBRUVICA was first approved by the U.S. Food and Drug Administration (FDA) in November 2013 and today is indicated for adult patients in six disease areas, including five hematologic cancers. These include indications to treat adults with chronic lymphocytic leukemia (CLL)/ small lymphocytic lymphoma (SLL) with or without 17p deletion (del17p); adults with Waldenström’s macroglobulinemia (WM); adult patients with previously treated mantle cell lymphoma (MCL)*; adult patients with previously treated marginal zone lymphoma (MZL) who require systemic therapy and have received at least one prior anti-CD20-based therapy*; and adult and pediatric patients aged one year and older with previously treated chronic graft-versus-host disease (cGVHD) after failure of one or more lines of systemic therapy.9

*Accelerated approval was granted for MCL and MZL based on overall response rate. Continued approval for MCL and MZL may be contingent upon verification and description of clinical benefit in confirmatory trials.

For more information, visit www.IMBRUVICA.com.

IMPORTANT SAFETY INFORMATION

WARNINGS AND PRECAUTIONS
Hemorrhage: Fatal bleeding events have occurred in patients who received IMBRUVICA. Major hemorrhage (≥ Grade 3, serious, or any central nervous system events; e.g., intracranial hemorrhage [including subdural hematoma], gastrointestinal bleeding, hematuria, and post procedural hemorrhage) occurred in 4.2% of patients, with fatalities occurring in 0.4% of 2,838 patients who received IMBRUVICA in 27 clinical trials. Bleeding events of any grade including bruising and petechiae occurred in 39%, and excluding bruising and petechiae occurred in 23% of patients who received IMBRUVICA, respectively.

The mechanism for the bleeding events is not well understood.

Use of either anticoagulant or antiplatelet agents concomitantly with IMBRUVICA increases the risk of major hemorrhage. Across clinical trials, 3.1% of 2,838 patients who received IMBRUVICA without antiplatelet or anticoagulant therapy experienced major hemorrhage. The addition of antiplatelet therapy with or without anticoagulant therapy increased this percentage to 4.4%, and the addition of anticoagulant therapy with or without antiplatelet therapy increased this percentage to 6.1%. Consider the risks and benefits of anticoagulant or antiplatelet therapy when co-administered with IMBRUVICA. Monitor for signs and symptoms of bleeding.

Consider the benefit-risk of withholding IMBRUVICA for at least 3 to 7 days pre- and post-surgery depending upon the type of surgery and the risk of bleeding.

Infections: Fatal and non-fatal infections (including bacterial, viral, or fungal) have occurred with IMBRUVICA therapy. Grade 3 or greater infections occurred in 21% of 1,476 patients who received IMBRUVICA in clinical trials. Cases of progressive multifocal leukoencephalopathy (PML) and Pneumocystis jirovecii pneumonia (PJP) have occurred in patients treated with IMBRUVICA. Consider prophylaxis according to standard of care in patients who are at increased risk for opportunistic infections. Monitor and evaluate patients for fever and infections and treat appropriately.

Cardiac Arrhythmias, Cardiac Failure, and Sudden Death: Fatal and serious cardiac arrhythmias and cardiac failure have occurred with IMBRUVICA. Deaths due to cardiac causes or sudden deaths occurred in 1% of 4,896 patients who received IMBRUVICA in clinical trials, including in patients who received IMBRUVICA in unapproved monotherapy or combination regimens. These adverse reactions occurred in patients with and without preexisting hypertension or cardiac comorbidities. Patients with cardiac comorbidities may be at greater risk of these events.

Grade 3 or greater ventricular tachyarrhythmias were reported in 0.2%, Grade 3 or greater atrial fibrillation and atrial flutter were reported in 3.7%, and Grade 3 or greater cardiac failure was reported in 1.3% of 4,896 patients who received IMBRUVICA in clinical trials, including in patients who received IMBRUVICA in unapproved monotherapy or combination regimens. These events have occurred particularly in patients with cardiac risk factors including hypertension and diabetes mellitus, a previous history of cardiac arrhythmias, and in patients with acute infections.

Evaluate cardiac history and function at baseline, and monitor patients for cardiac arrhythmias and cardiac function. Obtain further evaluation (e.g., ECG, echocardiogram) as indicated for patients who develop symptoms of arrhythmia (e.g., palpitations, lightheadedness, syncope, chest pain), new onset dyspnea, or other cardiovascular concerns. Manage cardiac arrhythmias and cardiac failure appropriately, follow dose modification guidelines, and consider the risks and benefits of continued IMBRUVICA treatment.

Hypertension: Hypertension occurred in 19% of 1,476 patients who received IMBRUVICA in clinical trials. Grade 3 or greater hypertension occurred in 8% of patients. Based on data from 1,124 of these patients, the median time to onset was 5.9 months (range, 0.03 to 24 months). Monitor blood pressure in patients treated with IMBRUVICA, initiate or adjust anti-hypertensive medication throughout treatment with IMBRUVICA as appropriate, and follow dosage modification guidelines for Grade 3 or higher hypertension.

Cytopenias: In 645 patients with B-cell malignancies who received IMBRUVICA as a single agent, grade 3 or 4 neutropenia occurred in 23% of patients, grade 3 or 4 thrombocytopenia in 8% and grade 3 or 4 anemia in 2.8%, based on laboratory measurements. Monitor complete blood counts monthly.

Second Primary Malignancies: Other malignancies (10%), including non-skin carcinomas (3.9%), occurred among the 1,476 patients who received IMBRUVICA in clinical trials. The most frequent second primary malignancy was non-melanoma skin cancer (6%).

Tumor Lysis Syndrome: Tumor lysis syndrome has been infrequently reported with IMBRUVICA. Assess the baseline risk (e.g., high tumor burden) and take appropriate precautions. Monitor patients closely and treat as appropriate.

Embryo-Fetal Toxicity: Based on findings in animals, IMBRUVICA can cause fetal harm when administered to a pregnant woman. Advise pregnant women of the potential risk to a fetus. Advise females of reproductive potential to use effective contraception during treatment with IMBRUVICA and for 1 month after the last dose. Advise males with female partners of reproductive potential to use effective contraception during the same time period.

ADVERSE REACTIONS

B-cell malignancies: The most common adverse reactions (≥30%) in adult patients with B-cell malignancies (MCL, CLL/SLL, WM and MZL) were thrombocytopenia (54.5%)*, diarrhea (43.8%), fatigue (39.1%), musculoskeletal pain (38.8%), neutropenia (38.6%)*, rash (35.8%), anemia (35.0%)*, and bruising (32.0%).

The most common Grade ≥ 3 adverse reactions (≥5%) in adult patients with B-cell malignancies (MCL, CLL/SLL, WM and MZL) were neutropenia (20.7%)*, thrombocytopenia (13.6%)*, pneumonia (8.2%), and hypertension (8.0%).

Approximately 9% (CLL/SLL), 14% (MCL), 14% (WM) and 10% (MZL) of adult patients had a dose reduction due to adverse reactions. Approximately 4-10% (CLL/SLL), 9% (MCL), and 7% (WM [5%] and MZL [13%]) of patients discontinued due to adverse reactions.

cGVHD: The most common adverse reactions (≥20%) in adult or pediatric patients with cGVHD were fatigue (57%), anemia (49%)*, bruising (40%), diarrhea (36%), thrombocytopenia (33%)*, musculoskeletal pain (30%), pyrexia (30%), muscle spasms (29%), stomatitis (29%), hemorrhage (26%), nausea (26%), abdominal pain (23%), pneumonia (23%), and headache (21%).

The most common Grade 3 or higher adverse reactions (≥5%) reported in adult or pediatric patients with cGVHD were pneumonia (14%), anemia (13%)*, fatigue (12%), pyrexia (11%), diarrhea (10%), neutropenia (10%)*, sepsis (10%), osteonecrosis (9%), stomatitis (9%), hypokalemia (7%), headache (5%), and musculoskeletal pain (5%).

Discontinuation of IMBRUVICA treatment due to an adverse reaction occurred in 24% of adult patients and 23% of pediatric patients. Adverse reactions leading to dose reduction occurred in 26% of adult patients and 19% of pediatric patients.

*Treatment-emergent decreases (all grades) were based on laboratory measurements.

DRUG INTERACTIONS

CYP3A Inhibitors: Co-administration of IMBRUVICA with strong or moderate CYP3A inhibitors may increase ibrutinib plasma concentrations. Increased ibrutinib concentrations may increase the risk of drug-related toxicity. Dose modifications of IMBRUVICA are recommended when used concomitantly with posaconazole, voriconazole, and moderate CYP3A inhibitors. Avoid concomitant use of other strong CYP3A inhibitors. Interrupt IMBRUVICA if strong inhibitors are used short-term (e.g., for ≤ 7 days). Avoid grapefruit and Seville oranges during IMBRUVICA treatment, as these contain strong or moderate inhibitors of CYP3A. See dose modification guidelines in USPI sections 2.3 and 7.1.

CYP3A Inducers: Avoid coadministration with strong CYP3A inducers.

SPECIFIC POPULATIONS

Pediatric Use: The safety and effectiveness of IMBRUVICA have not been established for the treatment of cGVHD after failure of one or more lines of therapy in pediatric patients less than 1 year of age. The safety and effectiveness of IMBRUVICA in pediatric patients have not been established in MCL, CLL/SLL, CLL/SLL with 17p deletion, WM, MZL or in patients with mature B-cell non-Hodgkin lymphoma.

In the randomized population from a study that included 35 patients (26 pediatric patients age 5 to less than 17 years) with previously treated mature B-cell non-Hodgkin lymphoma, major hemorrhage and discontinuation of chemoimmunotherapy due to adverse reactions occurred more frequently in the ibrutinib plus chemoimmunotherapy arm compared to the chemoimmunotherapy alone arm.

Hepatic Impairment:

Adult Patients with B-cell Malignancies: Hepatic Impairment (based on Child-Pugh criteria): Avoid use of IMBRUVICA in patients with severe hepatic impairment. In patients with mild or moderate impairment, reduce recommended IMBRUVICA dose and monitor more frequently for adverse reactions of IMBRUVICA.

Patients with cGVHD: Avoid use of IMBRUVICA in patients with total bilirubin level > 3x upper limit of normal (ULN) (unless of non-hepatic origin or due to Gilbert’s syndrome). Reduce recommended dose when administering IMBRUVICA to patients with total bilirubin level > 1.5 to 3x ULN (unless of non-hepatic origin or due to Gilbert’s syndrome).

Janssen Presents New Data for Talquetamab, a First-in-Class GPRC5DxCD3 Bispecific Antibody, Suggesting Durable Responses in Patients with Heavily Pretreated Multiple Myeloma

On December 10, 2022 The Janssen Pharmaceutical Companies of Johnson & Johnson reported updated results from the Phase 1/2 MonumenTAL-1 study of talquetamab, an investigational, off-the-shelf (ready to use), bispecific T-cell engager antibody (Press release, Janssen Biotech, DEC 10, 2022, View Source [SID1234625041]). Talquetamab targets both GPRC5D, a novel target on multiple myeloma cells, and CD3 on T cells, activating the body’s immune system to fight this blood cancer .1 Results from the study suggest patients with relapsed or refractory multiple myeloma who received a median of five prior lines of therapy, including a proteasome inhibitor, an immunomodulatory agent, and an anti-CD38 antibody, achieved overall response rates (ORR) of 74.1 percent and 73.1 percent with subcutaneously (SC) administered recommended Phase 2 doses (RP2Ds) of 0.4 mg/kg weekly and 0.8 mg/kg every two weeks, respectively, with a median duration of response (DOR) of nine months or longer.2 Data from this investigational trial were featured during the American Society of Hematology (ASH) (Free ASH Whitepaper) Annual Meeting as an oral presentation and highlighted in an ASH (Free ASH Whitepaper) press briefing on Saturday, December 10, 2022 (Abstract #157).2

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

"The responses observed in the MonumenTAL-1 study suggest the clinical potential of talquetamab in the treatment of many patients who are triple-class refractory," said Ajai Chari, M.D., Professor of Medicine, Hematology and Medical Oncology, Icahn School of Medicine, Mount Sinai, and principal study investigator.† "The results of this study indicate the potential of this treatment for heavily pretreated patients who have exhausted currently approved therapies."

Results from the Phase 1 portion of the MonumenTAL-1 study were also published in The New England Journal of Medicine on December 10, 2022.

The ORR to talquetamab treatment was similar across both dose schedules.2 With a median follow-up of 14.9 months (range 0.5+ to 29.0), 74.1 percent of patients treated at the SC 0.4 mg/kg dose administered weekly (QW) achieved a response, 59.4 percent achieved a very good partial response (VGPR) or better, 33.6 percent achieved a complete response (CR) or better and 23.8 percent achieved a stringent complete response (sCR).2 The median progression free survival (PFS) was 7.5 months (95 percent CI, 5.7–9.4) at the 0.4 mg/kg QW dose.2 At the SC 0.8 mg/kg dose administered every two weeks (Q2W), 73.1 percent of patients achieved a response, 57.2 percent achieved a VGPR or better, 32.4 percent achieved a CR or better, and 20 percent achieved a sCR at a median follow-up of 8.6 months (range 0.2 to 22.5).2 Due to shorter follow-up in the 0.8 mg/kg Q2W dose cohort, the median PFS is not yet mature.2 Median DOR was nine months or longer in all groups, and longer DOR was observed in those patients who achieved a CR or better.2 There were no significant differences in ORR in subgroup analysis including prior lines of therapy, refractoriness to prior therapy and cytogenetic risk at baseline.

"These data represent the importance of engaging T cells via CD3 and targeting GPRC5D – a tumor associated antigen which is overexpressed in multiple myeloma cells – for the treatment of patients with relapsed or refractory disease," said Sen Zhuang, M.D., Ph.D., Vice President, Clinical Research and Development, Janssen Research & Development, LLC. "Following our recent Biologics License Application submission to the U.S. FDA, we look forward to working with the agency to make this available as a treatment option in the short term and continuing our longer-term investigations of talquetamab as we aim to develop additional options for patients with this complex blood cancer."

An additional cohort was studied to determine response to talquetamab at either dose schedule after previous T-cell redirection therapy, either a CAR-T or CD3 bispecific antibody. Patients in this cohort were younger, had a higher prevalence of high-risk cytogenetics, and had received a median of six prior lines of therapy (range, 3 to 15).2 Among these patients, 70.6 percent received prior CAR-T-cell therapy and 35.3 percent received prior bispecific antibody therapy.2 At a median follow-up of 11.8 months (range 1 to 25.4), 62.7 percent of patients with prior T-cell redirection therapy achieved a response. An ORR of 72.2 percent (95 percent CI, 54.8–85.8) was observed in patients with prior CAR-T-cell therapy and 44.4 percent (95 percent CI, 21.5-69.2) in patients with prior bispecific antibody treatment.2 The safety profile was comparable in patients with and without prior T-cell redirection therapy.2

No new safety signals were identified with longer follow-up in Phase 1 or Phase 2 of the MonumenTAL-1 study.2 The most common adverse events (AEs) at the SC 0.4 mg/kg QW dose were cytokine release syndrome (CRS) (79 percent; 2.1 percent grade 3/4), skin-related AEs (55.9 percent; all grade 1/2) and nail-related AEs (51.7 percent; all grade 1/2).2 The most common AEs at the SC 0.8 mg/kg Q2W dose were CRS (72.4 percent; 0.7 percent grade 3/4), skin-related AEs (67.6 percent; 0.7 percent grade 3/4) and dysgeusia (46.2 percent; not applicable).2 CRS incidents were mostly grade 1/2 and largely confined to the step-up doses and first full dose.2 The incidence of infection was 57.3 percent at the SC 0.4 mg/kg QW dose and 50.3 percent at the SC 0.8 mg/kg Q2W dose, with 16.8 percent and 11.7 percent grade 3 or higher, respectively.2 One patient in each dose cohort died due to COVID-19 infection.2

Most high-grade hematologic AEs were cytopenias, which were generally limited to the first few cycles of treatment.2 Neutropenia was observed in 34.3 percent of patients treated at the SC 0.4 mg/kg QW dose (30.8 percent grade 3/4) and 28.3 percent of patients treated at the SC 0.8 mg/kg Q2W dose (22.1 percent grade 3/4).2 Thrombocytopenia was observed in 27.3 percent of patients treated at the SC 0.4 mg/kg QW dose (20.3 percent grade 3/4) and 26.9 percent of patients treated at the SC 0.8 mg/kg Q2W dose (16.6 percent grade 3/4).2 At the SC 0.4 mg/kg QW dose, 4.9 percent of patients discontinued treatment due to AEs; 8.4 percent had dose delays and 14.7 percent had dose reductions.2 At the SC 0.8 mg/kg Q2W dose, 6.2 percent of patients discontinued treatment due to AEs; 13.8 percent had dose delays and 6.2 percent had dose reductions.2

About Talquetamab

Talquetamab is a first-in-class, off-the-shelf (ready to use), investigational bispecific T-cell engager antibody targeting both GPRC5D, a novel multiple myeloma target, and CD3, a primary component of the T-cell receptor.2 CD3 is involved in activating T cells, and GPRC5D is highly expressed on multiple myeloma cells.3,4

Talquetamab, which is given by subcutaneous injection, is currently being evaluated in the Phase 1/2 MonumenTAL-1 clinical study for the treatment of relapsed or refractory multiple myeloma (NCT03399799), and in combination studies RedirecTT-1 (NCT04586426), TRIMM-2 (NCT04108195), TRIMM-3 (NCT05338775), MonumenTAL-2 (NCT05050097) and MonumenTAL-3 (NCT05455320).

On December 9, 2022, Janssen submitted a Biologics License Application to the U.S. Food and Drug Administration (FDA) for talquetamab for the treatment of patients with relapsed or refractory multiple myeloma. Talquetamab received Breakthrough Therapy designation from the U.S. FDA in June 2022 for the treatment of adult patients with relapsed or refractory multiple myeloma, who have previously received at least four prior lines of therapy, including a proteasome inhibitor, an immunomodulatory agent, and an anti-CD38 antibody. In May 2021 and August 2021, talquetamab was granted Orphan Drug designation for the treatment of multiple myeloma by the U.S. FDA and the European Commission, respectively. In January 2021, talquetamab received a PRIME designation by the European Commission.

About Multiple Myeloma

Multiple myeloma is an incurable blood cancer that affects a type of white blood cell called plasma cells, which are found in the bone marrow.5 In multiple myeloma, these plasma cells change, spread rapidly and replace normal cells in the bone marrow with tumors.6 In 2022, it is estimated that more than 34,000 people will be diagnosed with multiple myeloma, and more than 12,000 people will die from the disease in the U.S.7 While some people diagnosed with multiple myeloma initially have no symptoms, most patients are diagnosed due to symptoms that can include bone fracture or pain, low red blood cell counts, tiredness, high calcium levels, kidney problems or infections.