Sierra Oncology Reports Baseline Ferritin Differentially Predicts Week 24 Transfusion Independence Response in Myelofibrosis Patients

On December 13, 2021 Sierra Oncology, Inc. (NASDAQ: SRRA), a late-stage biopharmaceutical company on a mission to deliver targeted therapies that treat rare forms of cancer, reported data from new retrospective analyses of the Phase 3 SIMPLIFY studies demonstrate baseline ferritin differentially predicts Week 24 Transfusion Independence Response for momelotinib and ruxolitinib in patients with myelofibrosis (Press release, Sierra Oncology, DEC 13, 2021, View Source [SID1234596973]). The data were presented at the 63rd American Society of Hematology (ASH) (Free ASH Whitepaper) Annual Meeting being held virtually and in Atlanta, GA December 11-14, 2021.

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!

Baseline Serum Ferritin Differentially Predicts W24 Transfusion Independence Response for Momelotinib and Ruxolitinib in Patients with Myelofibrosis

Myelofibrosis is characterized by the presentation of constitutional symptoms, splenomegaly and anemia, with the degree of anemia and transfusion dependence being among the most important predictors of overall survival. Dr. Stephen Oh, MD, PhD, Washington University School of Medicine in St. Louis and Siteman Cancer Center, noted how the analyses identified pre-treatment serum ferritin level as the most predictive biomarker for the treatment effect of momelotinib versus ruxolitinib on Week 24 Transfusion Independence Response (TI-R) in the SIMPLIFY-1 (JAK inhibitor-naïve) and SIMPLIFY-2 (JAK-inhibitor experienced) studies.

Key results for SIMPLIFY-1 include:

Ruxolitinib was associated with a significantly greater increase in ferritin levels over time compared with momelotinib, irrespective of baseline ferritin, highlighting the differential treatment impact on serum ferritin between the two agents
Baseline hemoglobin (Hgb), an indicator of anemic status, differentially predicted Week 24 TI-R in patients randomized to momelotinib or ruxolitinib
Baseline ferritin also differentially predicted Week 24 TI-R in patients randomized to momelotinib or ruxolitinib
For patients with baseline ferritin between 90-650 ng/mL, momelotinib-treated patients had a higher Week 24 TI-R rate than ruxolitinib-treated patients [72% vs. 38%, OR 4.21 (95% CI: 2.24, 7.89); p=0.0439]
In patients with baseline Hgb <12 g/dL, baseline ferritin levels provided additional, differential predictive value for Week 24 TI-R
The differential treatment effect between momelotinib and ruxolitinib was highest for anemic patients with baseline ferritin between 90-650 ng/ml
No correlation was observed between baseline ferritin and Week 24 splenic or symptom response rates
Findings from SIMPLIFY-1 were independently confirmed in the JAK inhibitor experienced setting of the SIMPLIFY-2 study, where Hgb and ferritin each differentially predicted Week 24 TI-R in patients randomized to momelotinib or best available therapy (BAT; 88.5% ruxolitinib), and baseline ferritin predicted additional, differential predictive value for Week 24 TI-R in patients with Hgb <12 g/dL. For patients with baseline ferritin between 90-650 ng/mL, momelotinib-treated patients had higher Week 24 TI-R than those treated with BAT [53% vs 22%, OR 2.27 (95% CI: 1.01,12.77); p=0.03099].

These data suggest that ferritin may be useful in treatment decision making in myelofibrosis, especially in patients with anemia and ferritin 90-650 ng/mL, in which momelotinib demonstrates a greater TI effect than ruxolitinib. Future evaluation may be made in forthcoming clinical trials to further examine the correlation between ferritin and TI response.

About Momelotinib

Momelotinib is a selective and orally bioavailable JAK1, JAK2 and ACVR1/ALK2 inhibitor currently under investigation for the treatment of myelofibrosis. Myelofibrosis results from dysregulated JAK-STAT signaling and is characterized by constitutional symptoms, splenomegaly (enlarged spleen) and progressive anemia.

Sierra Oncology is currently awaiting topline results of the MOMENTUM clinical trial, a global, randomized, double-blind Phase 3 study evaluating momelotinib for the treatment of symptomatic and anemic myelofibrosis patients. Top-line data are anticipated by February 2022. Assuming positive data, the company plans to file a New Drug Application with the US Food & Drug Administration (FDA) in the second quarter of 2022. The FDA has granted Fast Track designation for momelotinib.

Atara Biotherapeutics Announces Positive Results from Pivotal Phase 3 Trial (ALLELE) of Tab-cel® at the 63rd American Society of Hematology (ASH) Annual Meeting

On December 13, 2021 Atara Biotherapeutics, Inc. (Nasdaq: ATRA), a leader in T-cell immunotherapy, leveraging its novel allogeneic Epstein-Barr virus (EBV) T-cell platform to develop transformative therapies for patients with cancer and autoimmune diseases, reported efficacy and safety results from its Phase 3 multicenter ALLELE study investigating tabelecleucel (tab-cel) for the treatment of Epstein-Barr virus positive post-transplant lymphoproliferative disease (EBV+ PTLD) following solid organ transplant (SOT) or hematopoietic cell transplant (HCT) (Press release, Atara Biotherapeutics, DEC 13, 2021, View Source [SID1234596972]). These findings, along with combined long-term survival data from Phase 2 and multicenter Expanded Access Protocol (EAP) studies of tab-cel were featured as oral presentations at the 63rd American Society of Hematology (ASH) (Free ASH Whitepaper) Annual Meeting.

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!

"Patients with EBV+ PTLD face a poor prognosis with survival measured in weeks to months if initial treatment is unsuccessful. There are no approved treatment options for this devastating disease, underscoring the critical unmet need that exists," said Jakob Dupont, MD, Head of Global Research & Development at Atara. "Our conviction that tab-cel, recently filed with the EMA, is a potential first-in-class treatment option for transplant recipients that develop EBV+ PTLD is validated by almost 90% of patients responding to treatment surviving after one year, and a similar two-year survival benefit of over 86% in patients who achieved a complete or partial response from Phase 2 and EAP studies."

Poor patient survival in relapsed or refractory EBV+ PTLD underscores the significant need for effective, safe, and fast-acting new therapeutic options as highlighted in two additional posters presented at ASH (Free ASH Whitepaper). Patients suffer from poor median survival of 0.7 months (n=81) and 4.1 months (n=86) for HCT and SOT, respectively, reported in EBV+ PTLD patients for whom rituximab ± chemotherapy failed.

In the ongoing Phase 3 ALLELE study, 38 evaluable patients as of May 2021 — 24 EBV+ PTLD patients following SOT after failure of rituximab ± chemotherapy and 14 EBV+ PTLD patients following HCT after failure of rituximab monotherapy — were treated with tab-cel and had the opportunity for a six-month follow-up after response. The median age of evaluable patients for both SOT and HCT was 52.9 years (3.2–81.5) who had tried a median of 1 (range: 1-5) prior systemic treatments including rituximab monotherapy, chemotherapy or immunotherapy.

As measured by independent oncologic response adjudication (IORA) assessment, an ORR of 50% (19/38, 95% CI: 33.4, 66.6) was observed for both HCT and SOT groups. For patients with EBV+ PTLD following SOT, an ORR of 50.0% (12/24, 95% CI: 29.1, 70.9) was observed and similarly, for patients with EBV+ PTLD following HCT, an ORR of 50.0% (7/14, 95% CI: 23.0, 77.0) was observed, with a best overall response of Complete Response (CR; 26.3%; n=10; n=5, SOT, n=5, HCT) or Partial Response (PR; 23.7%; n=9; n=7, SOT, n=2, HCT). The median time to response (TTR) in all patients was 1.1 months (0.7-4.7). In the study, 11 of 19 responders had a duration of response (DOR) lasting more than six months and median DOR has not yet been reached. Of the remaining eight responders, four had events due to IORA-assessed progressive disease (PD) or death and four patients were alive and censored for the DOR at the time of the data cut.

Patients responding to tab-cel had longer survival compared to the non-responders, with a median overall survival (OS) not evaluable (NE) (95% CI: 16.4, NE) and a one-year survival rate of 89.2% (95% CI: 63.1, 97.2) versus non-responders’ OS of 5.7 months (95% CI: 1.8, 12.1) and one-year survival rate of 32.4% (95% CI: 12.1, 54.9).

Safety was consistent with previously published data, and no new safety signals or concerns were reported. There were no reports of tumor flare reaction, infusion reactions, cytokine release syndrome, transmission of infectious diseases, including cytomegalovirus, and no events of graft versus host disease (GvHD) or organ rejection related to tab-cel. Overall, tab-cel was well-tolerated in treatment-refractory and immunocompromised patients.

Atara also reported combined long-term survival data from Phase 2 and multicenter EAP studies of tab-cel in a second oral presentation. Results show that across studies, patients who responded to tab-cel for treatment of EBV+ PTLD experienced a long-term survival benefit with a median OS of 54.6 months (95% CI: 14.8, 115.0) reported in all patients (n=76). An ORR of 63.2% (48/76) was observed in all patients with a best overall response of CR (42.1%; n=32) or PR (21.1%; n=16). Two-year survival rates were 86.2% (95% CI: 67.0, 94.6) and 86.5% (95% CI: 55.8, 96.5) for patients with CR and PR, respectively. Importantly, patients who achieved a PR with tab-cel derived similar OS benefit to those who achieved a CR. Treatment was well tolerated in refractory and immunocompromised patients and there were no fatal events reported related to tab-cel. There were no reports of tumor flare reaction, cytokine release syndrome, organ/marrow rejection, or transmission of infectious diseases and cytomegalovirus. There is no evidence of GvHD or infusion-related reaction risks attributable to tabelecleucel based on current data.

About Tabelecleucel

Tabelecleucel (tab‐cel) is an off-the-shelf, allogeneic T-cell immunotherapy in development for the treatment of Epstein-Barr virus-positive post-transplant lymphoproliferative disease (EBV+ PTLD). EBV+ PTLD is a type of lymphoma (cancer) that may occur after a solid organ transplant (SOT) or allogeneic hematopoietic cell transplant (HCT). There are currently no approved treatments indicated to treat PTLD and if left untreated, PTLD can have life-threatening consequences.

Tab-cel is currently being investigated in the Phase 3 registration-enabling ALLELE study to assess efficacy and safety for the treatment of EBV+ PTLD in SOT and HCT after failure of standard of care. These data support the recent EMA-validated Marketing Authorization Application for tab-cel as the first off-the-shelf allogeneic T-cell therapy ever to be reviewed by a regulatory agency. The EMA’s Committee for Medicinal Products for Human Use (CHMP) granted tab-cel Accelerated Assessment and an EU approval decision is anticipated for second half of 2022.

Tab-cel has been granted Breakthrough Therapy Designation for EBV+ PTLD following allogeneic HCT by the U.S. Food and Drug Administration (FDA) and PRIME designation by the European Medicines Agency (EMA) for the same indication. Tab-cel has orphan drug designation in the U.S. and EU.

Nanjing Leads Biolabs and BeiGene Announce Worldwide License and Collaboration Agreement for LBL-007 Anti-LAG-3 Antibody; BeiGene Granted Exclusive Commercialization Rights Outside of China

On December 13, 2021 BeiGene (NASDAQ: BGNE; HKEX: 06160), a global science-driven biotechnology company focused on developing innovative and affordable medicines to improve treatment outcomes and access for patients worldwide, and Nanjing Leads Biolabs, Inc. (Leads Biolabs), a privately-owned clinical stage biotechnology company in China and the U.S., reported entry into a license and collaboration agreement granting BeiGene worldwide research, development and manufacturing rights and exclusive commercialization rights outside of China to LBL-007, a novel investigational antibody targeting the LAG-3 pathway (Press release, BeiGene, DEC 13, 2021, View Source [SID1234596971]). Data from a Phase 1 clinical trial of LBL-007 in patients with advanced solid tumors were presented at the 2021 annual meeting of the American Society of Clinical Oncology (ASCO) (Free ASCO Whitepaper).

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!

"We are excited about the strategic opportunity of adding an anti-LAG-3 agent to our portfolio and the potential to expedite the clinical development and scientific understanding of both LBL-007 and the anti-LAG-3 pathway as monotherapy and in combination with other immuno-oncology assets in BeiGene’s portfolio, including our anti-PD-1 inhibitor tislelizumab, where we see exciting combination potential for improved anti-tumor activity," said Lai Wang, Ph.D., Global Head of R&D at BeiGene. "Nanjing Leads Biolabs has developed a promising clinical candidate, which complements our I/O program and supports our strategic imperatives of global clinical excellence and opportunities to address unmet medical needs around the world."

Under the terms of the agreement, Leads Biolabs will receive $30 million upfront and is eligible to receive up to $742 million in clinical development, regulatory approval, and sales milestones, plus tiered double-digit royalties on sales in the licensed territory.

"Securing a collaboration to further develop LBL-007 has been a key strategic priority, and we are excited to begin working with BeiGene, a global leader in oncology," said Xiaoqiang Kang, M.D., Ph.D., CEO and Chairman of Leads Biolabs. "BeiGene is the ideal partner for Leads Biolabs given its extensive experience in the development of oncology medicines worldwide and the compelling immuno-oncology combination opportunity in its pipeline. By collaborating with BeiGene, Leads Biolabs expects to significantly accelerate the development and commercialization of LBL-007."

About LBL-007
LAG-3 is an immune checkpoint receptor expressed on activated T cells to negatively regulate these cells, resulting in tumor immune escape. LBL-007, a novel investigational anti-LAG-3 antibody, was developed by screening of a human antibody phage display library and demonstrated specific binding to human LAG-3, stimulation of IL-2 release and blockage of LAG-3 binding to MHC II and other known LAG-3 ligands. LBL-007 monotherapy was shown in pre-clinical studies to significantly inhibit tumor growth, with more pronounced tumor inhibition when combined with an anti-PD-1 antibody. LBL-007 has obtained IND clearance in both the U.S. and China, as well as completed a Phase 1a clinical trial, and is currently in Phase 1b/2 clinical trials in China.

BeiGene Oncology
BeiGene is committed to advancing best and first-in-class clinical candidates internally or with like-minded partners to develop impactful and affordable medicines for patients across the globe. We have a growing R&D team of approximately 2,750 colleagues dedicated to advancing more than 90 ongoing or planned clinical trials (over 70 clinical trials are ongoing) involving more than 14,000 patients and healthy volunteers. Our expansive portfolio is directed predominantly by our internal colleagues supporting clinical trials in more than 45 countries and regions. Hematology-oncology and solid tumor targeted therapies and immuno-oncology are key focus areas for the Company, with both mono- and combination therapies prioritized in our research and development. BeiGene currently has three approved medicines discovered and developed in our own labs: BTK inhibitor BRUKINSA in the United States, China, the EU, Canada, Australia, and additional international markets; and the non-FC-gamma receptor binding anti-PD-1 antibody tislelizumab as well as the PARP inhibitor pamiparib in China.

BeiGene also partners with innovative companies who share our goal of developing therapies to address global health needs. We commercialize a range of oncology medicines in China licensed from Amgen, Bristol Myers Squibb, EUSA Pharma, and Bio-Thera. We also plan to address greater areas of unmet need globally through our collaborations including with Amgen, Mirati Therapeutics, Seagen, and Zymeworks. BeiGene has also entered into a collaboration with Novartis granting Novartis rights to develop, manufacture, and commercialize tislelizumab in North America, Europe, and Japan.

Gamida Cell Presents Two-Year Survival Data for GDA-201 and Resource Utilization Analysis for Omidubicel at 63rd ASH Annual Meeting

On December 13, 2021 Gamida Cell Ltd. (Nasdaq: GMDA), an advanced cell therapy company committed to cures for cancer and other serious diseases, reported that two-year follow-up data for GDA-201, the company’s lead candidate in its NAM-enabled NK cell therapy pipeline, will be presented at the 63rd American Society of Hematology (ASH) (Free ASH Whitepaper) Annual Meeting, being held in Atlanta, Georgia (Press release, Gamida Cell, DEC 13, 2021, View Source [SID1234596970]). Additionally, for patients who participated in the phase 3 trial of omidubicel, the company will be presenting a poster of an analysis of resource utilization data from the first 100 days after bone marrow transplant.

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 poster titled "GDA-201, A Novel Metabolically Enhanced Allogeneic Natural Killer (NK) Cell Product Yields High Remission Rates in Patients with Relapsed/Refractory Non-Hodgkin Lymphoma (NHL): 2-year survival and correlation with cytokine IL7" includes longer term follow-up from the phase 1, investigator-led study of GDA-201 in combination with rituximab (NCT03019666) in patients with relapsed or refractory non-Hodgkin lymphoma (NHL) and reports on 2-year outcomes and cytokine biomarkers associated with survival. The data demonstrated a median duration of response of 16 months (range 5- 36 months), an overall survival at 2 years of 78% (95% CI, 51%–91%) and a safety profile similar to that reported previously.

"We are excited to share this compelling two-year data from our investigator-led study of GDA-201 which demonstrate an extended duration of response in patients with NHL," said Julian Adams, Ph.D., Chief Executive Officer, of Gamida Cell. "The durable response in this patient group provides strong support as we work to progress GDA-201 through the development process for patients in need."

Gamida Cell will also present a poster related to its omidubicel program titled "Hospitalization and Healthcare Resource Use of Omidubicel Vs Cord Blood Transplantation for Hematological Malignancies in a Global Randomized Phase III Clinical Trial," which includes an analysis of resource utilization data from the first 100 days after transplant for 108 patients in the phase 3 trial showing that the omidubicel-treated patients had significantly shorter durations of hospitalization, intensive care unit stays, consultant visits, procedures, and transfusions than the control arm. These data provide further evidence of the clinical benefit associated with the more rapid hematopoietic recovery in patients treated with omidubicel and the corresponding reduction in healthcare resource utilization.

"This analysis clearly demonstrates the potential of omidubicel to significantly shorten a patient’s hospital length of stay, reduce time in ICU settings and decrease usage of healthcare resources, likely resulting in lower overall costs to the healthcare system," said Ronit Simantov, M.D., Chief Medical Officer of Gamida Cell. "These findings are particularly important as they demonstrate the impact of omidubicel on the experience for patients during the critical post-transplant period."

Both posters will be available today, Monday, December 13, 2021, 6:00-8:00 p.m. ET, during the ASH (Free ASH Whitepaper) Annual Meeting and Exposition.

About GDA-201

Gamida Cell applied the capabilities of its nicotinamide (NAM)-enabled cell expansion technology to develop GDA-201, an innate NK cell immunotherapy for the treatment of hematologic and solid tumors in combination with standard of care antibody therapies. GDA-201, the lead candidate in the NAM-enabled NK cell pipeline, has demonstrated promising initial clinical trial results. GDA-201 addresses key limitations of NK cells by increasing the cytotoxicity and in vivo retention and proliferation in the bone marrow and lymphoid organs. Furthermore, GDA-201 improves antibody-dependent cellular cytotoxicity (ADCC) and tumor targeting of NK cells. For more information about GDA-201, please visit View Source

GDA-201 is an investigational therapy, and its safety and efficacy have not been established by the FDA or any other health authority.

About Omidubicel

Omidubicel is an advanced cell therapy under development as a potential life-saving allogeneic hematopoietic stem cell (bone marrow) transplant solution for patients with blood cancers. Omidubicel is the first bone marrow transplant graft to receive Breakthrough Therapy Designation from the U.S. FDA and has also received Orphan Drug Designation in the U.S. and EU. For more information about omidubicel, please visit View Source

Omidubicel is an investigational therapy, and its safety and efficacy have not been established by the FDA or any other health authority.

Two-Year Analysis of CARTITUDE-1 Shows Early, Durable and Deepening Responses of Ciltacabtagene Autoleucel (cilta-cel) in Heavily Pretreated Patients with Multiple Myeloma

On December 13, 2021 Legend Biotech Corporation (NASDAQ: LEGN) (Legend Biotech), a global, clinical-stage biotechnology company developing and manufacturing novel therapies, reported that new and updated results from the CARTITUDE clinical development program studying ciltacabtagene autoleucel (cilta-cel) in the treatment of multiple myeloma, which were presented at the 63rd American Society of Hematology (ASH) (Free ASH Whitepaper) Annual Meeting and Exposition (Press release, Legend Biotech, DEC 13, 2021, View Source [SID1234596969]). Cilta-cel is an investigational B-cell maturation antigen (BCMA)-directed chimeric antigen receptor T cell (CAR-T) therapy being studied as a one-time treatment for multiple myeloma.

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!

CARTITUDE-1 Data Continues to Support the Potential of Cilta-cel

In an oral presentation (Abstract #549), longer-term results from the Phase 1b/2 CARTITUDE-1 study in 97 patients with relapsed or refractory multiple myeloma (RRMM) continued to show a very high overall response rate (ORR) of 98 percent. After 21.7 months of follow-up, 83 percent of patients treated with cilta-cel achieved a stringent complete response (sCR)—higher than the 67 percent sCR rate reported at a median of ~1 year of follow up.1 Further, 95 percent of patients achieved a very good partial response (VGPR) or better. Median progression-free survival (PFS) and median overall survival (OS) have not been reached, but the 2-year PFS rate was 61 percent (95 percent Confidence Interval [CI], 48.5–70.4) and the 2-year OS rate was 74 percent (95 percent CI, 61.9–82.7). Of the 61 patients evaluable for minimal residual disease (MRD), 92 percent were MRD-negative at the 10-5 cutoff threshold. The two-year PFS rates in patients with sustained MRD negativity for ≥6 and ≥12 months were 91 percent (95 percent CI, 67.1–97.8) and 100 percent, respectively.

The median time to first response was one month (range, 0.9-10.7); the median time to best response was 2.6 months (range, 0.9-17.8); and the median time to complete response or better was 2.9 months (range, 0.9-17.8).1 The longer-term data showed no new safety signals and there were no new events of cilta-cel-related neurotoxicity or movement and neurocognitive treatment emergent adverse events (TEAEs) (MNT) reported since the median ~1 year follow-up. Implementation of MNT mitigation measures has decreased the incidence rate to 0.5 percent in the CARTITUDE clinical development program.

In the 18-month follow-up data previously presented at ASCO (Free ASCO Whitepaper) 2021, the most common hematologic adverse events (AEs) observed were neutropenia (96 percent); anemia (81 percent); thrombocytopenia (79 percent); leukopenia (62 percent); and lymphopenia (53 percent).2 At 18 months, cytokine release syndrome (CRS) of any grade was observed in 95 percent of patients with a median duration of four days (range, 1-97), and median time to onset of seven days (range, 1-12). Of the 92 patients with CRS, 95 percent experienced Grade 1/2 events and CRS resolved in 91 patients (99 percent) within 14 days of onset. Neurotoxicity of any grade was observed in 21 percent (n=20) of patients, with Grade 3 or higher neurotoxicity observed in 10 percent (n=10) of patients.

"Patients with heavily pre-treated multiple myeloma often have exhausted available treatment options and face poor prognoses. The updated results from the CARTITUDE-1 trial continue to suggest that cilta-cel may provide this patient population with lasting deep and durable responses," said Thomas Martin, M.D., director of clinical research, clinical professor of medicine, Adult Leukemia and Bone Marrow Transplantation Program, interim Division Chief, co-director, Myeloma Program and co-leader, Hematopoietic Malignancies Program, at UCSF Helen Diller Family Comprehensive Cancer Center, and principal study investigator. "As a one-time infusion that shows potential to improve long-term survival and offer patients a break in ongoing treatments, cilta-cel may offer hope to patients, caregivers and physicians."

In a subgroup analysis of CARTITUDE-1 (Abstract #3938), responses to cilta-cel were durable up to 2 years in most subgroups of patients with heavily pretreated RRMM.3 An ORR range of 95 to 100 percent was observed in patients across all subgroups, including those with high-risk cytogenetics, International Staging System (ISS) stage III, baseline bone marrow cells ≥60 percent, and presence of baseline plasmacytomas. In patients with ISS stage III, high risk cytogenetics and with baseline plasmacytomas, median duration of response, 2-year PFS and OS appeared lower. The cilta-cel safety profile across the subgroups was consistent with the overall population, with no new safety signals.

Additionally, an adjusted indirect comparison of CARTITUDE-1 patient outcomes relative to standard-of-care therapies in real-world clinical practice (RWCP) was also featured in an oral presentation (Abstract #550).4 The adjusted comparisons versus CARTITUDE-1 demonstrate a significantly improved ORR, complete response or better (≥CR), VGPR or better (≥VGPR), PFS and OS for the patients receiving cilta-cel compared to a diverse set of RWCP. Although patients treated with cilta-cel experienced more adverse events (AEs), including Grade 3/4 events, as compared to RWCP, overall safety profile was manageable.

CARTITUDE-2 Data Explores Use of Cilta-cel in Earlier-Line MM Settings

The Phase 2 multicohort CARTITUDE-2 study is evaluating cilta-cel safety and efficacy in various clinical settings for patients with multiple myeloma. Updated data from Cohort A of the study examined the efficacy and safety of cilta-cel in 20 patients with progressive multiple myeloma after 1-3 prior lines of therapy and who are lenalidomide-refractory (Abstract #3866).5 At a longer median follow-up of 14.3 months, patients experienced early and deep responses with a manageable safety profile consistent with the CARTITUDE-1 study. ORR was 95 percent, which included 85 percent of patients achieving CR or better and 90 percent achieving VGPR or better. The median time to first response was one month (range, 0.7-3.3) and the median time to best response was 2.6 months (range, 0.9-7.9). The 6-month and 12-month PFS rates were 95 percent (95 percent CI, 69.5-99.3) and 84 percent (95 percent CI, 59.1-94.7), respectively. Of the 13 patients with MRD evaluable samples at the 10-5 cutoff threshold, 92 percent (95 percent CI, 64.0-99.8) were MRD negative.

The first data from Cohort B was also presented at ASH (Free ASH Whitepaper) 2021 (Abstract #2910).6 Cohort B included 19 patients who were in early relapse after initial therapy that included a proteasome inhibitor (PI) and immunomodulatory drug (IMiD). Data showed early and deep responses with a manageable safety profile. At a median follow-up of 10.6 months, ORR was 95 percent, which included 79 percent of patients achieving CR or better and 90 percent of patients achieving VGPR or better. The median time to first response was one month (range, 0.9-2.6) and the median time to best response was 2.5 months (range, 0.9-11.8). The 6-month and 12-month PFS rates were 90 percent (95 percent CI, 64.1-97.3) and 84 percent (95 percent CI, 57.9-94.5), respectively. Of the 13 patients with MRD evaluable samples at the 10-5 cutoff threshold, 92 percent (95 percent CI, 64.0-99.8) were MRD-negative.

The safety profile seen in CARTITUDE-2 Cohorts A and B were consistent with data previously reported from CARTITUDE-1. CRS occurred in 95 percent of patients in Cohort A and 84 percent of patients in Cohort B, which were mostly grades 1/2 with median time to onset of 7-8 days and median duration of ~4 days.

"The new and updated longer-term data for CARTITUDE-1 and Cohorts A and B of CARTITUDE-2 shows that responses continue to be deep and durable over time and illustrate the potential of cilta-cel to provide a new treatment option for those patients that need it the most," said Ying Huang, PhD, CEO and CFO of Legend Biotech. "We are excited to continue to present these strong efficacy and safety results as we work toward the first regulatory approval for cilta-cel and from our robust cell therapy pipeline."

About CARTITUDE-1

CARTITUDE-1 (NCT03548207) is a Phase 1b/2, open-label, multicenter study evaluating the safety and efficacy of cilta-cel in adults with relapsed and/or refractory with multiple myeloma who have received at least 3 prior lines of therapy or are double refractory to a proteasome inhibitor (PI) and immunomodulatory drug (IMiD), received a PI, an IMiD, and anti-CD38 antibody and documented disease progression within 12 months of starting the most recent therapy. The primary objective of the Phase 1b portion of the study was to characterize the safety and confirm the recommended Phase 2 dose of cilta-cel, informed by the first-in-human study with LCAR-B38M CAR-T cells (LEGEND-2). The Phase 2 portion further evaluated the efficacy of cilta-cel with overall response rate as the primary endpoint.

About CARTITUDE-2

CARTITUDE-2 (NCT04133636) is an ongoing Phase 2 multicohort study evaluating the safety and efficacy of cilta-cel in various clinical settings. Cohort A included patients who had progressive multiple myeloma after 1–3 prior lines of therapy, including PI and IMiD, were lenalidomide refractory, and had no prior exposure to BCMA-targeting agents. Cohort B included patients with early relapse after initial therapy that included a PI and IMiD. The primary objective was percentage of patients with negative minimal residual disease (MRD).

About LocoMMotion

LocoMMotion (NCT04035226) is a prospective non-interventional study evaluating the safety and efficacy of real-life standard-of-care treatments under routine clinical practice over a 24-month period in patients with RRMM. This study aims to understand the effectiveness of current standards of care in heavily pretreated patients with RRMM (reflecting real-world practice in the patient population progressing after PIs, IMiDs and anti-CD38 antibodies).

About Multiple Myeloma

Multiple myeloma is an incurable blood cancer that starts in the bone marrow and is characterized by an excessive proliferation of plasma cells.7 Although treatment may result in remission, unfortunately, patients will most likely relapse.8 Relapsed myeloma is when the disease has returned after a period of initial, partial or complete remission and does not meet the definition of being refractory.9 Refractory multiple myeloma is when a patient’s disease is non-responsive or progresses within 60 days of their last therapy.10,11 While some patients with multiple myeloma have no symptoms at all, most patients are diagnosed due to symptoms that can include bone problems, low blood counts, calcium elevation, kidney problems or infections.12 Patients who relapse after treatment with standard therapies, including protease inhibitors and immunomodulatory agents, have poor prognoses and few treatment options available.13

About Cilta-cel

Cilta-cel is an investigational chimeric antigen receptor T cell (CAR-T) therapy, formerly identified as JNJ-4528 in the United States and Europe and LCAR-B38M CAR-T cells in China, that is being studied in a comprehensive clinical development program for the treatment of patients with relapsed or refractory multiple myeloma and in earlier lines of treatment. The design consists of a structurally differentiated CAR-T with two BCMA-targeting single domain antibodies. In December 2017, Legend Biotech, Inc. entered into an exclusive worldwide license and collaboration agreement with Janssen Biotech, Inc. (Janssen) to develop and commercialize cilta-cel. In addition to a Breakthrough Therapy Designation (BTD) granted in the United States in December 2019, cilta-cel received a Priority Medicines (PRiME) designation from the European Commission in April 2019, and a BTD in China in August 2020. In addition, Orphan Drug Designation was granted for cilta-cel by the U.S. Food and Drug Administration (FDA) in February 2019, and by the European Commission in February 2020. A Biologics License Application seeking approval of cilta-cel was submitted to the U.S. FDA and a Marketing Authorization Application was submitted to the European Medicines Agency.