Jazz Pharmaceuticals Presents Positive Interim Phase 2/3 Results of Rylaze™ (asparaginase erwinia chrysanthemi (recombinant)-rywn) in Acute Lymphoblastic Leukemia or Lymphoblastic Lymphoma at ASH 2021 Annual Meeting

On December 11, 2021 Jazz Pharmaceuticals plc (Nasdaq: JAZZ) reported initial positive results from a Phase 2/3 trial of intramuscular (IM) administration of Rylaze (asparaginase erwinia chrysanthemi (recombinant)-rywn) in adult and pediatric patients with acute lymphoblastic leukemia (ALL) and lymphoblastic lymphoma (LBL) who have developed hypersensitivity or silent inactivation to an E. coli-derived asparaginase (Press release, Jazz Pharmaceuticals, DEC 11, 2021, View Source [SID1234596882]). The study was developed and conducted in close collaboration with the Children’s Oncology Group (COG). These initial results will be presented for the first time today 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!

Three cohorts with unique, IM administration dosing schedules were evaluated in the trial, demonstrating a safety profile consistent with other asparaginases. In Cohort 1c, a dosing regimen of Rylaze administered 25 mg/m2 on Monday and Wednesday and 50 mg/m2 on Friday demonstrated a positive benefit-to-risk profile, showing that Rylaze maintains a clinically meaningful level of nadir serum asparaginase activity (NSAA) ≥0.1 IU/mL at both 48 and 72 hours. Rylaze was approved by the U.S. Food and Drug Administration (FDA) on June 30, 2021 under the Real-Time Oncology Review (RTOR) program for use as a component of a multi-agent chemotherapeutic regimen for the treatment of ALL or LBL in adult and pediatric patients one month and older who have developed hypersensitivity to E. coli-derived asparaginase. Rylaze was approved at the dosing schedule of 25 mg/m2 every 48 hours based on data from Cohort 1a, in conjunction with data produced by a preliminary population pharmacokinetic (PPK) model.

These data will support additional regulatory filings for Rylaze, including a supplemental Biologics Licensing Application (sBLA) in early 2022 for a Monday/Wednesday/Friday (M/W/F) IM dosing schedule that will be reviewed under the FDA RTOR program. These data will also support regulatory submissions in Europe in mid-2022, with potential for approval in 2023.

"Asparaginase is an integral part of ALL therapy that is associated with improvement in survival rates. Following FDA approval earlier this year, Rylaze is already providing patients who have developed hypersensitivity to E. coli-derived asparaginase with a much-needed, effective therapeutic option with reliable supply and consistently high quality," said Rob Iannone, M.D., M.S.C.E., executive vice president, research and development and chief medical officer of Jazz Pharmaceuticals. "Rylaze is proof of Jazz’s ability to take medicines from concept through development, approval and launch, and we look forward to working with the FDA through the sBLA submission with these additional data in early 2022 in support of a label expansion for M/W/F dosing."

"The results from the Phase 2/3 study for Rylaze help to expand our knowledge of its dosing and safety profile, and support Monday/Wednesday/Friday dosing, which is more in line with clinical practice," said primary study investigator Dr. Luke Maese, associate professor at the University of Utah, Primary Children’s Hospital and Huntsman Cancer Institute. "The accelerated development and approval of Rylaze ensured that many patients with LBL and ALL – most of whom are children – who cannot tolerate E. coli-derived asparaginases have a new treatment option that maintains therapeutic levels of asparaginase activity throughout duration of treatment."

Interim Trial Results
Data presented at ASH (Free ASH Whitepaper) 2021 include initial analyses from an ongoing Phase 2/3 open-label, multicenter, dose confirmation and pharmacokinetic (PK) study of Rylaze (also known as JZP458) in patients with ALL/LBL who developed hypersensitivity or silent inactivation to a long-acting E. coli-derived asparaginase. Preliminary data are from Part A of the study, which investigated three Cohorts via IM administration:

Cohort 1a (n=33): studied a dose of 25 mg/m2 Monday/Wednesday/Friday
Cohort 1b (n=53): studied a dose of 37.5 mg/m2 Monday/Wednesday/Friday
Cohort 1c (n=52): studied a dose of 25 mg/m2 on Monday and Wednesday and 50 mg/m2 on Friday
Part B of the Phase 2/3 study remains active to further confirm the dose and schedule of the intravenous (IV) route of administration for Rylaze.

Efficacy Findings
The primary efficacy endpoints of the trial were the proportion of patients with a last 72-hour (from Friday to Monday) NSAA levels of ≥0.1 IU/mL during the first treatment course, in addition to safety and tolerability of Rylaze in patients with ALL/LBL.

The key secondary endpoint included the proportion of patients achieving the last 48-hour NSAA ≥0.1 IU/mL during the first treatment course.

The proportion of patients with observed NSAA levels ≥0.1 IU/mL with a 95% CI during Course 1 from these initial results is as follows (primary and key secondary endpoints):

Cohort 1a

Cohort 1b

Cohort 1c

At 48 hours

97% (CI: 91%, 100%)

98% (CI: 95%, 100%)

96% (CI: 90%, 100%)

At 72 hours

66% (CI: 48%, 83%)

80% (CI: 70%, 91%)

90% (CI:81%, 98%)

Based on a PPK modeling and simulation analysis versus observed data for Cohort 1c, the proportion of patients predicted to achieve NSAA levels ≥0.1 IU/mL with a 95% CI from these initial results is as follows:

Observed

Model Prediction

At 48 hours

96% (CI: 90%, 100%)

93% (CI: 92%, 94%)

At 72 hours

90% (CI:81%, 98%)

91% (CI: 90%, 92%)

The mean serum asparaginase activity (SAA) levels were also determined: mean SAA levels (95% CIs) from the initial data in Cohorts 1a, 1b and 1c at 48 hrs were 0.45 IU/mL (0.37, 0.53), 0.84 IU/mL (0.68, 0.99), and 0.66 IU/mL (0.54, 0.77); and at 72 hrs were 0.15 IU/mL (0.12, 0.19), 0.30IU/mL (0.23, 0.37), and 0.46 IU/mL (0.34, 0.58), respectively. These results reflect the higher dose on Friday from Cohort 1c.

Safety Findings
Grade 3/4 treatment-emergent adverse events (TEAEs), regardless of causality, occurred in 78/137 (57%) patients. There were no treatment-related TEAEs leading to death. The most commonly reported non-hematologic TEAEs (in ≥20% in any cohort) regardless of causality included: vomiting, nausea, fatigue, decreased appetite, pyrexia, abdominal pain, alanine aminotransferase (ALT) increased, febrile neutropenia, back pain, headache, sinus tachycardia, stomatitits, pain in extremity, aspartate aminotransferase (AST) increased and hyperglycemia. Treatment-related TEAEs leading to study drug discontinuation occurred in 6/137 (4%) of patients.

Overall, the safety profile of Rylaze was consistent with the reported safety information for patients with ALL/LBL receiving asparaginase with combination chemotherapy.

Further study analyses (including PK and safety analyses) are ongoing, and full study results will be reported at a later date.

About Rylaze (asparaginase erwinia chrysanthemi (recombinant)-rywn)
Rylaze, also known as JZP458, is approved in the U.S. for use as a component of a multi-agent chemotherapeutic regimen for the treatment of acute lymphoblastic leukemia (ALL) and lymphoblastic lymphoma (LBL) in adult and pediatric patients one month or older who have developed hypersensitivity to E. coli-derived asparaginase. Rylaze has orphan drug designation for the treatment of ALL/LBL in the United States. Rylaze is a recombinant erwinia asparaginase that uses a novel Pseudomonas fluorescens expression platform. JZP458 was granted Fast Track designation by the U.S. Food and Drug Administration (FDA) in October 2019 for the treatment of this patient population. Rylaze was approved as part of the Real-Time Oncology Review (RTOR) program, an initiative of the FDA’s Oncology Center of Excellence designed for efficient delivery of safe and effective cancer treatments to patients.

The full U.S. Prescribing Information for Rylaze is available at: View Source

Important Safety Information

RYLAZE should not be given to people who have had:

Serious allergic reactions to RYLAZE
Serious swelling of the pancreas (stomach pain), serious blood clots, or serious bleeding during previous asparaginase treatment
RYLAZE may cause serious side effects, including:

Allergic reactions (a feeling of tightness in your throat, unusual swelling/redness in your throat and/or tongue, or trouble breathing), some of which may be life-threatening
Swelling of the pancreas (stomach pain)
Blood clots (may have a headache or pain in leg, arm, or chest)
Bleeding
Liver problems
Contact your doctor immediately if any of these side effects occur.

Some of the most common side effects with RYLAZE include: liver problems, nausea, bone and muscle pain, tiredness, infection, headache, fever, allergic reactions, fever with low white blood cell count, decreased appetite, mouth swelling (sometimes with sores), bleeding, and too much sugar in the blood.

RYLAZE can harm your unborn baby. Inform your doctor if you are pregnant, planning to become pregnant, or nursing. Females of reproductive potential should use effective contraception (other than oral contraceptives) during treatment and for 3 months following the final dose. Do not breastfeed while receiving RYLAZE and for 1 week after the final dose.

Tell your healthcare provider if there are any side effects that are bothersome or that do not go away.

These are not all the possible side effects of RYLAZE. For more information, ask your healthcare provider.

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 (1-800-332-1088).

About Acute Lymphoblastic Leukemia (ALL)
ALL is a cancer of the blood and bone marrow that can progress quickly if not treated.1 Leukemia is the most common cancer in children, and about three out of four of these cases are ALL.2 Although it is one of the most common cancers in children, ALL is among the most curable of the pediatric malignancies due to recent advancements in treatment.3,4 Adults can also develop ALL, and about four of every 10 cases of ALL diagnosed are in adults.4 The American Cancer Society estimates that almost 6,000 new cases of ALL will be diagnosed in the United States in 2021.4 Asparaginase is a core component of multi-agent chemotherapeutic regimens in ALL.5 However, asparaginase treatments derived from E. coli are associated with the potential for development of hypersensitivity reactions.6

About Lymphoblastic Lymphoma (LBL)
LBL is a rare, fast-growing, aggressive subtype of Non-Hodgkin’s lymphoma, most often seen in teenagers and young adults.6 LBL is a very aggressive lymphoma – also called high-grade lymphoma – which means the lymphoma grows quickly with early spread to different parts of the body.[7],[8]

Ascentage Pharma Announces CDE’s Approval for the Phase II Pivotal Study in China of the Bcl-2 Inhibitor Lisaftoclax (APG-2575) for the Treatment of Relapsed/Refractory Chronic Lymphocytic Leukemia/Small Lymphocytic Lymphoma

On December 11, 2021 Ascentage Pharma (6855.HK), a global biopharmaceutical company engaged in developing novel therapies for cancers, chronic hepatitis B (CHB), and age-related diseases, reported that the Phase II pivotal study (the APG2575CC201 study) of the company’s novel Bcl-2 selective inhibitor, lisaftoclax (APG-2575), for the treatment of relapsed/refractory chronic lymphocytic leukemia/small lymphocytic lymphoma (R/R CLL/SLL) has been approved by the Center for Drug Evaluation (CDE) in China (Press release, Ascentage Pharma, DEC 11, 2021, View Source [SID1234596878]).

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!

APG2575CC201 is a single-arm, open-label, Phase II pivotal study designed to evaluate the efficacy and safety of lisaftoclax, with objective response rate (ORR) as the primary endpoint. Based on existing safety and efficacy data of lisaftoclax, the CDE has agreed that results from the APG2575CC201 study can be used to support the future New Drug Application for the indication of R/R CLL/SLL.

Lisaftoclax is a novel, orally administered Bcl-2 selective inhibitor being developed by Ascentage Pharma. Lisaftoclax is designed to treat a variety of malignancies by selectively blocking Bcl-2 to restore the normal apoptosis process in cancer cells. It is the first China-developed Bcl-2 inhibitor entering clinical development in China. Lisaftoclax is being studied in multiple clinical studies in countries and regions including the U.S., China, Australia, and the European Union, for a range of hematologic malignancies and solid tumors such as CLL/SLL. At the 2021 American Society of Hematology (ASH) (Free ASH Whitepaper) Annual Meeting, data of China studies of lisaftoclax in the treatment of hematologic malignancies were released for the first time. These data demonstrated lisaftoclax’s favorable tolerability and enormous therapeutic potential, without evidence of any tumor lysis syndrome (TLS). The six patients with CLL who received lisaftoclax at 200 mg or higher doses achieved an ORR of 100% and one case of complete response (CR).

Globally, there are significant unmet medical needs in the treatment of CLL/SLL. Patients with R/R CLL/SLL, especially those who are refractory or resistant to immunotherapies, chemotherapies, and Bruton Tyrosine Kinase (BTK) inhibitors, commonly experience rapid disease progression and currently lack any effective treatment, thus represents an urgent need for an effective novel therapy.

"Lisaftoclax is a key candidate in our apoptosis-targeted pipeline. In earlier studies, it has demonstrated promising efficacy and safety implicating great best-in-class potential. The approval for this Phase II pivotal study makes us even more confident in the developmental and regulatory pathway of lisaftoclax," said Dr. Yifan Zhai, Chief Medical Officer of Ascentage Pharma. "There are urgent unmet medical needs in patients with R/R CLL/SLL who currently lack effective salvage therapies. We will actively advance this Phase II pivotal study of lisaftoclax to hopefully offer a new treatment option to patients around the world, and let more patients with CLL/SLL who now lack any effective treatment benefit from this novel therapeutic as soon as possible."

About Lisaftoclax (APG-2575)
Lisaftoclax is a novel, orally administered small-molecule Bcl-2‒selective inhibitor being developed by Ascentage Pharma to treat hematologic malignancies and solid tumors by selectively blocking antiapoptotic protein Bcl-2 and hence restoring the normal apoptosis process in cancer cells. Lisaftoclax is the first China-developed Bcl-2 inhibitor entering clinical development in China.

Lisaftoclax is being studied in multiple clinical studies in countries and regions including the U.S., China, Australia, and the European Union, for a range of hematologic malignancies and solid tumors such as chronic lymphocytic leukemia, acute myeloid leukemia, and breast cancer. Lisaftoclax has been granted Orphan Drug Designations for five indications including Waldenström macroglobulinemia, chronic lymphocytic leukemia, multiple myeloma, acute myeloid leukemia, and follicular lymphoma.

Valemetostat Pivotal Data Shows Promising Response Rates in Patients with Adult T-Cell Leukemia/Lymphoma

On December 11, 2021 Daiichi Sankyo Company, Limited (hereafter, Daiichi Sankyo) reported that valemetostat, a potential first-in-class dual inhibitor of EZH1 and EZH2, demonstrated promising response rates in a pivotal phase 2 study in Japanese patients with relapsed/refractory adult T-cell leukemia/lymphoma (ATL) (Press release, Daiichi Sankyo, DEC 11, 2021, View Source [SID1234596865]). The results were reported today in an oral presentation (#303) at the 63rd Annual Meeting of the American Society of Hematology (ASH) (Free ASH Whitepaper) (#ASH21).

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!

While ATL is a rare disease, it occurs with greater frequency in parts of Japan and other regions.1 Currently, there are no optimal standard treatment options for ATL.1 Nearly 90% of patients relapse after completing intensive first-line treatment, at which point there are few options available.1,2

The phase 2 study of valemetostat met its primary endpoint, demonstrating an objective response rate (ORR) of 48% (90% CI: 30.5%-65.9%) in 25 patients with relapsed/refractory ATL, as evaluated by an independent efficacy assessment committee. Five complete remissions, seven partial remissions and 10 cases of stable disease were reported. The median duration of response (DOR) was not reached (95% CI: 1.87 months-NR) at a median follow-up of 6.5 months. Eight patients remained on treatment at the time of data cut-off on April 24, 2021.

The safety profile of valemetostat in the study was consistent with the phase 1 trial in patients with several types of non-Hodgkin lymphoma including peripheral T-cell lymphoma (PTCL) and ATL.3 Grade 3 or higher treatment emergent adverse events (TEAEs) occurred in 15 of 25 patients (60%), the most common of which (occurring in ≥30% of patients) were platelet count decrease (80%), anemia (48%), alopecia (40%) and dysgeusia (36%). Dose interruptions or reductions due to TEAEs occurred in 20% (n=5) and 8% (n=2) of patients, respectively. Two patients (8%) discontinued treatment due to TEAEs.

Based on these data, valemetostat was granted Orphan Drug designation by the Japan Ministry of Health, Labour and Welfare (MHLW) for the treatment of relapsed/refractory ATL. Therapies receiving Orphan Drug designation from the Japan MHLW are those being developed for serious, difficult-to-treat diseases that affect fewer than 50,000 patients in Japan, and they qualify for several measures intended to support development including, but not limited to, guidance and subsidies for research and development activities, priority consultation for clinical development and priority review of applications.

"The phase 2 trial of valemetostat demonstrated encouraging response rates in Japanese patients with a history of mogamulizumab therapy for relapsed/refractory ATL," said Makoto Yoshimitsu, MD, PhD, Associate Professor, Kagoshima University Hospital, Japan. "For patients in Japan with aggressive ATL subtypes, median overall survival is about 12 months even with intensive chemotherapy regimens, and potential new options such as valemetostat are greatly needed to improve outcomes, particularly in the relapsed/refractory setting."4

"Based on these pivotal data for valemetostat, dual targeting of EZH1 and EZH2, which play important roles in the pathophysiology of T-cell lymphomas, appears to be a promising therapeutic approach for patients with relapsed/refractory ATL, a type of PTCL with particularly poor prognosis," said Ken Takeshita, MD, Global Head of R&D, Daiichi Sankyo. "We are working to deliver valemetostat to ATL patients in Japan as soon as possible while continuing global development in T-cell and B-cell lymphomas."

The study also showed a trend towards a decrease in measurable lesions across all disease sites assessed, including nodal, extranodal, skin and peripheral blood.

The trial included patients with three aggressive subtypes of ATL who received a median of three prior lines of therapy (range, 1-8). Twenty-four of 25 patients had received prior treatment with mogamulizumab.

Summary of Phase 2 Results

Efficacy Measure*

All Patients

(n=25)


Acute

(n=16)


Lymphoma

(n=6)


Unfavorable chronic

(n=3)

ORR, n (%)

12 (48.0)


10 (62.5)


1 (16.7)


1 (33.3)

CR

5 (20.0)


5 (31.3)


0


0

CRu

0


0


0


0

PR

7 (28.0)


5 (31.3)


1 (16.7)


1 (33.3)

SD

10 (40.0)


4 (25.0)


5 (83.3)


1 (33.3)

RD/PD

3 (12.0)


2 (12.5)


0


1 (33.3)

Median time to first response (months)

1.43

(range 1.0-5.6)


n/a


n/a


n/a

Median DOR (months)

NR

(95% CI: 1.87-NR)


n/a


n/a


n/a

*CR, complete remission; CRu, complete remission unconfirmed; DOR, duration of response; NR, not reached; ORR, objective response rate; PD, progressive disease; PR, partial response; RD, relapsed disease; SD, stable disease

About Adult T-Cell Leukemia/Lymphoma

Adult T-cell leukemia/lymphoma (ATL) is a rare and aggressive type of peripheral T-cell lymphoma (PTCL) that is caused by human T-cell lymphotropic virus type 1 (HTLV-1).1 More than 3,000 new cases of ATL are diagnosed each year worldwide.5 While ATL is rare, it occurs with greater frequency in some regions including parts of Japan, the Caribbean, South America and Australia.5 In Japan, there are approximately 1,000 new ATL cases and over 1,000 deaths due to ATL annually.6

ATL continues to have a dismal prognosis with current therapies.7 The five-year overall survival rate for patients with ATL is about 14%.8 A median survival time of approximately eight months (252 days) was reported for patients in Japan with the most common acute ATL subtype.5

Treatment of ATL is based on subtype and consists primarily of intensive multi-drug chemotherapy regimens.7 Nearly 90% of patients relapse after completing intensive first-line treatment, at which point there are few options available.1,2 Additional therapies are needed to improve the prognosis of ATL in Japan and worldwide.1,7

About EZH1 and EZH2

EZH1 (enhancer of zeste homolog 1) and EZH2 (enhancer of zeste homolog 2) enzymes are part of polycomb protein complexes and act through histone methylation to regulate expression of genes involved in maintaining hematopoietic stem cells.9 EZH1 and EZH2 are recurrently highly expressed or mutated in many hematologic malignancies including T-cell lymphomas.10 Research has demonstrated that both EZH1 and EZH2 enzymes have a role in hematologic cancer progression and that simultaneous inhibition would be effective in targeting the cancers.11 There are no dual inhibitors of EZH1 and EZH2 approved for cancer treatment.

About the Phase 2 Study

The pivotal, open-label, multi-center, single-arm phase 2 study evaluated efficacy and safety of valemetostat (200 mg dose daily) as monotherapy in patients with relapsed/refractory ATL who were previously treated with mogamulizumab or at least one systemic chemotherapy in case of intolerance/contraindication for mogamulizumab and with no history of allogenic hematopoietic stem cell transplant.

The primary endpoint is ORR assessed by independent efficacy assessment committee. Secondary endpoints include investigator-assessed ORR, best response in tumor lesions, complete remission rate, tumor control rate, time to response, duration of response, progression-free survival, overall survival and safety. A total of 25 patients were enrolled in the study in Japan. For more information, visit ClinicalTrials.gov.

About Valemetostat

Valemetostat is a potential first-in-class dual inhibitor of EZH1 and EZH2 currently in clinical development in the Alpha portfolio of Daiichi Sankyo. A potent and selective small molecule inhibitor, valemetostat is designed to counter epigenetic dysregulation by targeting both the EZH1 and EZH2 enzymes.12

The valemetostat development program includes VALENTINE-PTCL01, a global pivotal phase 2 trial in patients with relapsed/refractory PTCL and ATL; a pivotal phase 2 trial in patients with relapsed or refractory ATL in Japan; and, a phase 1 study in patients with relapsed/refractory NHL in the U.S. and Japan. Valemetostat received Orphan Drug Designation (ODD) from the U.S. Food & Drug Administration for the treatment of PTCL in December 2021, ODD from the Japan MHLW for the treatment of relapsed/refractory ATL in November 2021 and SAKIGAKE Designation from the Japan MHLW for the treatment of adult patients with relapsed/refractory PTCL in April 2019.

Valemetostat is an investigational medicine that has not been approved for any indication in any country. Safety and efficacy have not been established.

About Daiichi Sankyo Oncology

The oncology portfolio of Daiichi Sankyo is powered by our team of world-class scientists that push beyond traditional thinking to create transformative medicines for people with cancer. Anchored by our DXd antibody drug conjugate (ADC) technology, our research engines include biologics, medicinal chemistry, modality and other research laboratories in Japan, and Plexxikon Inc., our small molecule structure-guided R&D center in the U.S. We also work alongside leading academic and business collaborators to further advance the understanding of cancer as Daiichi Sankyo builds towards our ambitious goal of becoming a global leader in oncology by 2025.

BeiGene Presents Results from SEQUOIA Trial of BRUKINSA (zanubrutinib) in First-Line Chronic Lymphocytic Leukemia at the 63rd ASH Annual Meeting

On December 11, 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, reported results from a planned interim analysis of the Phase 3 SEQUOIA trial in patients with treatment-naïve (TN) chronic lymphocytic leukemia (CLL), including the randomized Cohort 1 comparing BRUKINSA to bendamustine plus rituximab (B+R) and Cohort 3 (Arm D) of BRUKINSA in combination with venetoclax in patients with deletion of chromosome 17p (del[17p]) and/or pathogenic TP53 variants (Press release, BeiGene, DEC 11, 2021, View Source [SID1234596858]). These data were reported in two oral presentations at the 63rd American Society for 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!

"In the positive SEQUOIA trial, BRUKINSA delivered the therapeutic promise of a selective BTK inhibitor as a frontline treatment for CLL patients, with demonstrated superiority over chemoimmunotherapy. These robust data, along with the results from our previously reported Phase 3 ALPINE trial, strengthen our belief that BRUKINSA could become an important new treatment option for patients with CLL," remarked Jane Huang, M.D., Chief Medical Officer of Hematology at BeiGene. "In addition, BRUKINSA achieved favorable safety advantages in both trials such as lower rates of atrial fibrillation."

"Compared to chemoimmunotherapy, BRUKINSA demonstrated superior PFS benefit for CLL patients receiving frontline treatment, including those harboring high-risk characteristics, such as unmutated IGHV status and del(11q)," said Constantine Tam, MBBS, M.D., Peter MacCallum Cancer Center, and a principal investigator of the study. "Safety findings in SEQUOIA were similar to what has been reported in other BRUKINSA clinical trials, with consistently low rates of atrial fibrillation. Based on these results, BRUKINSA, as a highly selective BTK inhibitor, can potentially provide a chemo-free treatment option for CLL patients."

For more information on BeiGene’s clinical program and company updates, please visit BeiGene’s virtual booth at this year’s ASH (Free ASH Whitepaper) Annual Meeting at View Source

SEQUOIA Cohort 1: BRUKINSA vs. B+R in TN CLL Patients Without del (17p)

Oral Presentation; Abstract #396; Plain language summary available at View Source

A total of 479 patients with TN CLL whose tumor did not exhibit del(17p) were enrolled in Cohort 1 of the SEQUOIA trial, with 241 patients randomized to receive BRUKINSA (Arm A) and 238 patients randomized to receive B+R (Arm B). Patient characteristics were balanced between the two arms, with more than 50% with unmutated IGHV gene and 18% with del(11q) in each. Patients with del(17p) typically have poor response to chemoimmunotherapy and were assigned to receive BRUKINSA treatment in Cohort 2. Results from Cohort 2 were previously presented at the 2020 ASH (Free ASH Whitepaper) Annual Meeting.

The primary endpoint of the SEQUOIA trial is progression-free survival (PFS) per independent review committee (IRC) assessment in the randomized Cohort 1.

At the interim analysis, with a median follow-up of 26.15 months, BRUKINSA demonstrated superiority in PFS over B+R, as assessed by IRC. Results included:

The 24-month PFS rate was 85.5% (95% CI: 80.1, 89.6) in Arm A, compared to 69.5% (95% CI: 62.4, 75.5) in Arm B, with a hazard ratio (HR) of 0.42 (95% CI: 0.27, 0.63), p < 0.0001;
PFS benefit was consistently observed across key patient subgroups, including patients with del(11q), unmutated IGHV status, Binet stage C, and bulky disease; and
Overall survival (OS) results were early, and at 24 months, OS probability was similar between two arms, with 94.3% (95% CI: 90.4, 96.7) in Arm A and 94.6% (95% CI: 90.6, 96.9) in Arm B.
Safety analysis was based on 240 patients in Arm A and 227 patients in Arm B who received at least one dose of respective treatment. BRUKINSA was generally well tolerated with a safety profile consistent with its broad clinical program, including a low rate of atrial fibrillation. Results included:

224 patients (93.3%) in Arm A experienced at least one adverse event (AE) of any grade, with the most common (≥12%) being contusion (19.2%), upper respiratory tract infection (17.1%), neutropenia (15.4%), diarrhea (13.8%), and arthralgia (13.3%);
In comparison, 218 patients (96.0%) in Arm B experienced at least one AE of any grade, with the most common (≥12%) being neutropenia (56.8%), nausea (32.6%), pyrexia (26.4%), rash (19.4%), anemia (18.9%), constipation (18.9%), infusion-related reaction (18.9%), fatigue (15.9%), vomiting (14.5%), thrombocytopenia (13.7%), and diarrhea (13.2%);
126 patients (52.5%) in Arm A experienced at least one Grade ≥3 AE, compared to 181 patients (79.7%) in Arm B, with the most common in both arms being neutropenia (11.3% in Arm A vs. 51.1% in Arm B) and thrombocytopenia (1.7% in Arm A vs. 7.0% in Arm B);
88 patients (36.7%) in Arm A experienced at least one serious AE, compared to 113 patients (49.8%) in Arm B;
AEs leading to dose reduction, interruption or delay, and discontinuation occurred in 18 patients (7.5%), 111 patients (46.3%), and 20 patients (8.3%), respectively, in Arm A, compared to 84 patients (37.4%), 154 patients (67.8%), and 31 patients (13.7%), respectively, in Arm B;
Fatal AEs were reported in 11 patients (4.6%) in Arm A, compared to 11 patients (4.8%) in Arm B;
AEs of interest of any grade included anemia (Arm A vs. Arm B: 4.6% vs. 19.4%), arthralgia (13.3% vs. 8.8%), atrial fibrillation (3.3% vs. 2.6%), bleeding (45.0% vs. 11.0%), diarrhea (13.8% vs. 13.7%), hypertension (14.2% vs. 10.6%), infections (62.1% vs. 55.9%), myalgia (3.8% vs. 1.3%), neutropenia (15.8% vs. 56.8%), other cancers (12.9% vs. 8.8%), and thrombocytopenia (4.6% vs. 17.6%).
In addition, efficacy results with an extended follow-up from Cohort 2 (Arm C) of BRUKINSA as a monotherapy in patients with del(17p) were reported at ASH (Free ASH Whitepaper). With a median follow-up of 30.5 months, the 24-month PFS rate was 88.9% (95% CI: 81.3, 93.6).

Summary of SEQUOIA Cohort 1 Interim Analysis

SEQUOIA Cohort 1

Summary


BRUKINSA

(n=241)

Bendamustine + Rituximab

(n=238)

Efficacy Results

IRC-Assessed

24-month PFS (Primary Endpoint)


85.5%

(95% CI: 80.1, 89.6)

69.5%

(95% CI: 62.4, 75.5)


Hazard Ratio=0.42 (95%CI: 0.27, 0.63)

2-sided p <0.0001

Overall Safety Results

AEs of any grade


93.3%

96.0%

Grade ≥3 AEs


52.5%

79.7%

Serious AEs


36.7%

49.8%

AEs leading to dose reduction


7.5%

37.4%

AEs leading to dose interruption or delay


46.3%

67.8%

AEs leading to treatment discontinuation


8.3%

13.7%

Fatal AEs


4.6%

4.8%

Adverse Events of Interest (Any Grade)

Anemia


4.6%

19.4%

Neutropenia


15.8%

56.8%

Thrombocytopenia


4.6%

17.6%

Arthralgia


13.3%

8.8%

Atrial fibrillation


3.3%

2.6%

Bleeding


45.0%

11.0%

Diarrhea


13.8%

13.7%

Hypertension


14.2%

10.6%

Infections


62.1%

55.9%

Myalgia


3.8%

1.3%

Other cancers


12.9%

8.8%

SEQUOIA Cohort 3 (Arm D): BRUKINSA + Venetoclax in TN CLL Patients with del(17p) and/or TP53 Mutations

Oral Presentation; Abstract #67

Cohort 3 of SEQUOIA was designed to examine the hypothesis that the addition of venetoclax to BRUKINSA can drive tumors into deeper remission. Building on the demonstrated efficacy and safety of BRUKINSA in Cohort 2, Cohort 3 is planned to enroll approximately 80 patients with TN CLL whose tumor exhibits del(17p) or TP53 mutations, with key endpoints being safety, overall response rate (ORR), PFS, and duration of response (DoR). These patients will receive BRUKINSA treatment at 160 mg twice daily for three months, followed by combination treatment of BRUKINSA at the same dosing and venetoclax with a ramp-up dosing to 400 mg once daily for 12 to 24 cycles until progressive disease, unacceptable toxicity, or confirmed undetectable measurable residual disease (uMRD).

"Unfavorable prognosis is often seen in CLL patients with del(17p) or pathogenic TP53 variants, even in the front-line setting. While the follow-up in Cohort 3 was relatively short, the high response rate and the deepened responses observed among those treated for longer periods suggested the potential of BRUKINSA in combination with venetoclax in these high-risk CLL patients. The combination treatment also appeared generally well tolerated," commented Alessandra Tedeschi, M.D., Grande Ospedale Metropolitano Niguarda in Italy, a principal investigator on the study. "We look forward to the continued evaluation of BRUKINSA in combination with venetoclax in untreated CLL patients with del(17p) or TP53 mutations."

At the data cutoff on September 7, 2021, 49 patients were enrolled in Cohort 3, including 46 patients (93.9%) with centrally confirmed positive del(17p) status and three patients (6.1%) with a pathogenic TP53 variant alone. Patients enrolled in Cohort 3 also exhibited other markers of high risk, including 87.8% with unmutated IGHV, 91.9% with concurrent TP53 mutation, and 83.3% with complex karyotype (at least three abnormalities).

With a short median follow-up of 12.0 months, a high ORR was observed in the 36 patients who had at least one post-baseline response evaluation by the data cutoff date. Preliminary efficacy results per investigator assessment included:

Of the 14 patients who received combination treatment for more than 12 months, five patients (36%) achieved a confirmed complete response (CR) or CR with incomplete bone marrow recovery (CRi) in a bone marrow assessment and four additional patients met the criteria for CR or CRi but not confirmed in bone marrow assessment due to COVID-19 restrictions; and
In all 36 patients evaluable for efficacy, the ORR was 97.2% (95% CI: 85.5, 99.9) and the CR/CRi rate was 13.9% (all CRs or CRis were in patients who received combination treatment for more than 12 months).
With a median follow-up of 7.9 months, safety results in all 49 enrolled patients included:

40 patients (81.6%) experienced at least one AE of any grade, with the most common (≥12%) being infections (16.3%), neutropenia (14.3%), bruising (12.2%), diarrhea (12.2%), minor bleeding (12.2%), and nausea (12.2%);
16 patients (32.7%) experienced at least one Grade ≥3 AE and four patients (8.2%) experienced at least one serious AE;
AEs leading to dose interruption, dose reduction, and treatment discontinuation occurred in 10 patients (20.4%), no patients (0.0%), and one patient (2.0%), respectively; and
One patient (2.0%) experienced a fatal AE.
With a median follow-up of 13.5 months, safety results in the 34 patients who received combination treatment included:

29 patients (85.3%) experienced at least one AE of any grade, with the most common (≥12%) being infections (23.5%), neutropenia (20.6%), diarrhea (14.7%), fatigue (14.7%), nausea (14.7%), and bruising (11.8%);
13 patients (38.2%) experienced at least one Grade ≥3 AE and three patients (8.8%) experienced at least one serious AE; and
AEs leading to dose interruption occurred in 10 patients (29.4%), with no AEs leading to dose reduction or treatment discontinuation.
About SEQUOIA

SEQUOIA is a randomized, multicenter, global Phase 3 trial (NCT03336333) designed to evaluate the efficacy and safety of BRUKINSA compared to B+R in patients with TN CLL or SLL. The trial consists of three cohorts:

Cohort 1 (n=479): randomized 1:1 to receive BRUKINSA (n=241) or B+R (n=238) until disease progression or unacceptable toxicity, in patients not harboring del(17p); data from this group comprise the primary endpoint;
Cohort 2 (n=110): patients with del(17p) receiving BRUKINSA as a monotherapy; and
Cohort 3 (enrollment ongoing): patients with del(17p) or pathogenic TP53 variant receiving BRUKINSA in combination with venetoclax.
Patients with del(17p) were not randomized to B+R, as they experience poor clinical outcomes and poor response to chemoimmunotherapy. The primary endpoint of the trial is IRC-assessed PFS. Secondary endpoints include investigator-assessed PFS, IRC- and investigator-assessed overall response rate (ORR), overall survival (OS), PFS and ORR in patients with del(17p), and safety.

Cohort 2 (Arm C), representing high-risk patients treated with BRUKINSA monotherapy, was previously presented at the American Society for Hematology (ASH) (Free ASH Whitepaper) Annual Meeting in December 2020. This cohort of patients with del(17p) achieved significant efficacy with an 18-month PFS of 90.6%, as assessed by investigator.

About Chronic Lymphocytic Leukemia and Small Lymphocytic Lymphoma

Chronic lymphocytic leukemia (CLL) is the most common form of leukemia in adults, with a global incidence of approximately 114,000 new cases in 2017.1,2 CLL affects white blood cells or lymphocytes in the bone marrow.1 Proliferation of cancer cells (leukemia) in the marrow result in reduced ability to fight infection and spread into the blood, which affects other parts of the body including the lymph nodes, liver and spleen.1,3 The BTK pathway is a known route that signals malignant B cells and contributes to the onset of CLL.4 Small lymphocytic lymphoma (SLL) is a non-Hodgkin’s lymphoma affecting the B-lymphocytes of the immune system, which shares many similarities to CLL but with cancer cells found mostly in lymph nodes.5

About BRUKINSA

BRUKINSA is a small molecule inhibitor of Bruton’s tyrosine kinase (BTK) discovered by BeiGene scientists that is currently being evaluated globally in a broad clinical program as a monotherapy and in combination with other therapies to treat various B-cell malignancies. Because new BTK is continuously synthesized, BRUKINSA was specifically designed to deliver complete and sustained inhibition of the BTK protein by optimizing bioavailability, half-life, and selectivity. With differentiated pharmacokinetics compared to other approved BTK inhibitors, BRUKINSA has been demonstrated to inhibit the proliferation of malignant B cells within a number of disease relevant tissues.

BRUKINSA has received 12 approvals covering 40 countries and regions:

For the treatment of mantle cell lymphoma (MCL) in adult patients who have received at least one prior therapy (United States, November 2019)*;
For the treatment of MCL in adult patients who have received at least one prior therapy (China, June 2020)**;
For the treatment of chronic lymphocytic leukemia (CLL) or small lymphocytic lymphoma (SLL) in adult patients who have received at least one prior therapy (China, June 2020)**;
For the treatment of relapsed or refractory MCL (United Arab Emirates, February 2021);
For the treatment of Waldenström’s macroglobulinemia (WM) in adult patients (Canada, March 2021);
For the treatment of adult patients with WM who have received at least one prior therapy (China, June 2021)**;
For the treatment of MCL in adult patients who have received at least one prior therapy (Canada, July 2021);
For the treatment of MCL in adult patients who have received at least one prior therapy (Chile, July 2021);
For the treatment of adult patients with MCL who have received at least one previous therapy (Brazil, August 2021);
For the treatment of adult patients with WM (United States, August 2021);
For the treatment of adult patients with marginal zone lymphoma (MZL) who have received at least one anti-CD20-based regimen (United States, September 2021)*;
For the treatment of adult patients with MCL who have received at least one previous therapy (Singapore, October 2021);
For the treatment of MCL in patients who have received at least one prior therapy (Israel, October 2021);
For the treatment of adult patients with WM who have received at least one prior therapy, or in first line treatment for patients unsuitable for chemo-immunotherapy (Australia, October 2021);
For the treatment of adult patients with MCL who have received at least one prior therapy (Australia, October 2021);
For the treatment of adult patients with MCL who have received at least one previous therapy (Russia, October 2021);
For the treatment of adult patients with MCL who have received at least one previous therapy (Saudi Arabia, November 2021); and
For the treatment of adult patients with WM who have received at least one prior therapy or first-line treatment of patients unsuitable for chemo-immunotherapy (European Union plus Iceland and Norway, November 2021).
To date, more than 20 marketing authorization applications have been submitted for BRUKINSA for various indications.

* This indication was approved under accelerated approval based on overall response rate. Continued approval for this indication may be contingent upon verification and description of clinical benefit in a confirmatory trial.

** This indication was approved under conditional approval. Complete approval for this indication may be contingent upon results from ongoing randomized, controlled confirmatory clinical trials.

IMPORTANT U.S. SAFETY INFORMATION FOR BRUKINSA (ZANUBRUTINIB)

Warnings and Precautions

Hemorrhage

Fatal and serious hemorrhagic events have occurred in patients with hematological malignancies treated with BRUKINSA monotherapy. Grade 3 or higher hemorrhage including intracranial and gastrointestinal hemorrhage, hematuria and hemothorax have been reported in 3.4% of patients treated with BRUKINSA monotherapy. Hemorrhage events of any grade occurred in 35% of patients treated with BRUKINSA monotherapy.

Bleeding events have occurred in patients with and without concomitant antiplatelet or anticoagulation therapy. Co-administration of BRUKINSA with antiplatelet or anticoagulant medications may further increase the risk of hemorrhage.

Monitor for signs and symptoms of bleeding. Discontinue BRUKINSA if intracranial hemorrhage of any grade occurs. Consider the benefit-risk of withholding BRUKINSA for 3-7 days pre- and post-surgery depending upon the type of surgery and the risk of bleeding.

Infections

Fatal and serious infections (including bacterial, viral, or fungal) and opportunistic infections have occurred in patients with hematological malignancies treated with BRUKINSA monotherapy. Grade 3 or higher infections occurred in 27% of patients, most commonly pneumonia. Infections due to hepatitis B virus (HBV) reactivation have occurred.

Consider prophylaxis for herpes simplex virus, pneumocystis jiroveci pneumonia and other infections according to standard of care in patients who are at increased risk for infections. Monitor and evaluate patients for fever or other signs and symptoms of infection and treat appropriately.

Cytopenias

Grade 3 or 4 cytopenias, including neutropenia (26%), thrombocytopenia (11%) and anemia (8%) based on laboratory measurements, developed in patients treated with BRUKINSA monotherapy. Grade 4 neutropenia occurred in 13% of patients, and Grade 4 thrombocytopenia occurred in 3.6% of patients.

Monitor complete blood counts regularly during treatment and interrupt treatment, reduce the dose, or discontinue treatment as warranted. Treat using growth factor or transfusions, as needed.

Second Primary Malignancies

Second primary malignancies, including non-skin carcinoma, have occurred in 14% of patients treated with BRUKINSA monotherapy. The most frequent second primary malignancy was non-melanoma skin cancer, reported in 8% of patients. Other second primary malignancies included malignant solid tumors (4.0%), melanoma (1.7%) and hematologic malignancies (1.2%). Advise patients to use sun protection and monitor patients for the development of second primary malignancies.

Cardiac Arrhythmias

Atrial fibrillation and atrial flutter were reported in 3.2% of patients treated with BRUKINSA monotherapy. Patients with cardiac risk factors, hypertension, and acute infections may be at increased risk. Grade 3 or higher events were reported in 1.1% of patients treated with BRUKINSA monotherapy. Monitor signs and symptoms for atrial fibrillation and atrial flutter and manage as appropriate.

Embryo-Fetal Toxicity

Based on findings in animals, BRUKINSA can cause fetal harm when administered to a pregnant woman. Administration of zanubrutinib to pregnant rats during the period of organogenesis caused embryo-fetal toxicity including malformations at exposures that were 5 times higher than those reported in patients at the recommended dose of 160 mg twice daily. Advise women to avoid becoming pregnant while taking BRUKINSA and for 1 week after the last dose. Advise men to avoid fathering a child during treatment and for 1 week after the last dose.

If this drug is used during pregnancy, or if the patient becomes pregnant while taking this drug, the patient should be apprised of the potential hazard to a fetus.

Adverse reactions

The most common adverse reactions, including laboratory abnormalities, in ≥ 30% of patients who received BRUKINSA (N = 847) included decreased neutrophil count (54%), upper respiratory tract infection (47%), decreased platelet count (41%), hemorrhage (35%), decreased lymphocyte count (31%), rash (31%) and musculoskeletal pain (30%).

Drug Interactions

CYP3A Inhibitors: When BRUKINSA is co-administered with a strong CYP3A inhibitor, reduce BRUKINSA dose to 80 mg once daily. For coadministration with a moderate CYP3A inhibitor, reduce BRUKINSA dose to 80 mg twice daily.

CYP3A Inducers: Avoid coadministration with moderate or strong CYP3A inducers.

Specific Populations

Hepatic Impairment: The recommended dose of BRUKINSA for patients with severe hepatic impairment is 80 mg orally twice daily.

Please see full U.S. Prescribing Information at www.beigene.com/PDF/BRUKINSAUSPI.pdf and Patient Information at www.beigene.com/PDF/BRUKINSAUSPPI.pdf.

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 that have involved 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 and Bristol Myers Squibb. We also plan to address greater areas of unmet need globally through our collaborations including with Amgen, Bio-Thera, EUSA Pharma, 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.

New Data from CARTITUDE-1 Study Show Continued Deep and Durable Responses of Ciltacabtagene Autoleucel (cilta-cel) in Treatment of Heavily Pre-treated Patients with Multiple Myeloma

On December 11, 2021 The Janssen Pharmaceutical Companies of Johnson & Johnson reported longer-term results from the Phase 1b/2 CARTITUDE-1 study evaluating the efficacy and safety of ciltacabtagene autoleucel (cilta-cel), an investigational B-cell maturation antigen (BCMA)-directed chimeric antigen receptor T-cell (CAR-T) therapy administered as a single infusion, in the treatment of patients with relapsed and/or refractory multiple myeloma (Press release, Johnson & Johnson, DEC 11, 2021, View Source [SID1234596856]).1 The data, featured as an oral presentation at the American Society of Hematology (ASH) (Free ASH Whitepaper) 2021 Annual Meeting (Abstract #549) and selected as part of the Highlights of ASH (Free ASH Whitepaper) programme, show that patients receiving cilta-cel demonstrate deep and durable responses, with a very high overall response rate (ORR) of 98 percent.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!

Responses in the 97 patients treated with cilta-cel deepened over time, with 83 percent of patients achieving a stringent complete response (sCR) at median 22-month follow-up, an increase from 80 percent at the 18-month median follow-up data presented at the 2021 American Society of Clinical Oncology (ASCO) (Free ASCO Whitepaper) Annual Meeting, and from 67 percent at the 12.4-month median follow-up data presented at ASH (Free ASH Whitepaper) 2020.1,2,3 At median follow-up of 22 months, median progression-free survival (PFS) and median overall survival (OS) were not reached, suggesting long-term durability of responses and survival for patients.1 Two-year PFS and OS rates were 61 percent (95 percent Confidence Interval [CI], 48.5–70.4) and 74 percent (95 percent CI, 61.9–82.7), respectively.1 Among 61 minimal residual disease (MRD) evaluable patients, 92 percent of patients achieved MRD negativity (at 10-5).1 The two-year PFS rates in patients who achieved MRD negativity for ≥six and ≥12 months were 91 percent (95 percent CI, 67.1–97.8) and 100 percent, respectively.1

"Unfortunately, patients with multiple myeloma for whom at least three treatment regimens have stopped working, face a median survival of less than a year with currently available treatments," said Thomas Martin, M.D.,* Director of Clinical Research, Clinical Professor of Medicine, Adult Leukemia and Bone Marrow Transplantation Program, Associate Director, Myeloma Program and Co-Leader, Hematopoietic Malignancies Program, at UCSF Helen Diller Family Comprehensive Cancer Center, and principal study investigator. "The CARTITUDE-1 data presented at ASH (Free ASH Whitepaper) 2021 builds upon previous results that show that a single infusion of cilta-cel resulted in durable responses and survival across the study population, further supporting the potential of cilta-cel in offering patients and physicians a valuable new treatment option."

Median time to first response was one month (range, 0.9–10.7), with responses deepening over time.1 Additionally, median time to best response was 2.6 months (range, 0.9–17.8) and median time to complete response or better was 2.9 months (range, 0.9–17.8).1 Twelve percent of patients achieved a very good partial response and three percent achieved a partial response.1 The study included patients who received a median of six prior treatment regimens (range, 3–18).1 All patients were triple-class [immunomodulatory agent (IMiD), proteasome inhibitor (PI) and an anti-CD38 antibody] exposed, while 42 percent of patients were penta-drug refractory and 99 percent of patients were refractory to the last line of therapy.1

"These data add to the growing body of evidence supporting the potential clinical benefit of cilta-cel in the treatment of patients with relapsed and/or refractory multiple myeloma, a population in need of new options," said Sen Zhuang, M.D., Ph.D., Vice President, Clinical Research and Development, Janssen Research & Development, LLC. "We look forward to further evaluation of cilta-cel as part of our comprehensive CARTITUDE clinical development programme that includes studying cilta-cel in patients with newly diagnosed multiple myeloma."

The data demonstrated a consistent safety profile for cilta-cel and no new safety signals were observed with longer follow-up.1 In 18-month follow-up data presented at ASCO (Free ASCO Whitepaper) 2021, the most common haematologic adverse events (AEs) observed were neutropenia (96 percent); anaemia (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 99 percent of which resolved within 14 days of onset.2 Of the 92 patients with CRS at 18-month follow-up, 95 percent experienced grade 1/2 events.2 Neurotoxicity of any grade was observed in 21 percent (n=20) of patients at 18 months, with grade 3 or higher neurotoxicity observed in 10 percent (n=10) of patients.2 There were no new events of cilta-cel–related neurotoxicity or movement and neurocognitive treatment (MNT) emergent adverse events reported in CARTITUDE-1 since the median 12.4-month follow-up data were presented at ASH (Free ASH Whitepaper) 2020.1,3 At the 22-month data cut-off, more than 200 patients have been dosed with cilta-cel across the CARTITUDE clinical development programme and MNT incidence has decreased to less than one percent since the implementation of MNT mitigation measures.1

"We are pleased that we are again able to present longer-term follow-up data, this time at nearly two years, for this innovative potential treatment for people living with relapsed/refractory multiple myeloma." said Edmond Chan MBChB M.D. (Res), EMEA Therapeutic Area Lead Haematology, Janssen-Cilag Limited. "The sustained efficacy and consistent safety profile build on the strong results seen to date and may bring us one step closer to changing the expectations of what a multiple myeloma diagnosis means for patients."

Subgroup Analysis of the Phase 1b/2 CARTITUDE-1 Study
In the abstract accepted for presentation at ASH (Free ASH Whitepaper) 2021, data demonstrated that cilta-cel resulted in deep, durable responses in all evaluated subgroups in CARTITUDE-1 at median follow-up of 18 months.4 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 multiple myeloma, baseline bone marrow cells ≥60 percent, and presence of baseline plasmacytomas.4 In patients with ISS stage III, high risk cytogenetics and with baseline plasmacytomas, median duration of response appeared shorter and 18-month PFS and OS rates lower.4 The cilta-cel safety profile across the subgroups was consistent with the overall population, with no new safety signals.4 The latest data from this analysis will be presented in a poster presentation (Abstract #3938) at ASH (Free ASH Whitepaper) 2021 on Monday, December 13.4

# ENDS #

About CARTITUDE-1
CARTITUDE-1 (NCT03548207)5 is an ongoing Phase 1b/2, open-label, multicentre study evaluating the safety and efficacy of cilta-cel in adults with relapsed and/or refractory multiple myeloma, 99 percent of whom were refractory to the last line of treatment; 88 percent of whom were triple-class refractory, meaning their cancer did not respond, or no longer responds, to an immunomodulatory agent (IMiD), a proteasome inhibitor (PI) and an anti-CD38 antibody.1

The primary objective of the Phase 1b portion of the study, involving 29 patients, was to characterise the safety and confirm the dose of cilta-cel, informed by the first-in-human study with LCAR-B38M CAR-T cells (LEGEND-2).1,5,6 Based on the safety profile observed in this portion of the study, outpatient dosing is being evaluated in additional CARTITUDE studies. The Phase 2 portion of the study, involving 68 additional patients, is evaluating the efficacy of cilta-cel with overall response as the primary endpoint.1,5

About Ciltacabtagene Autoleucel (cilta-cel)
Cilta-cel is an investigational chimeric antigen receptor T-cell (CAR-T) therapy that is being studied in a comprehensive clinical development programme for the treatment of patients with relapsed or refractory multiple myeloma and in earlier lines of treatment.1 The design consists of a structurally differentiated CAR-T with two BCMA-targeting single domain antibodies.1 In December 2017, Janssen Biotech, Inc. (Janssen) entered into an exclusive worldwide license and collaboration agreement with Legend Biotech to develop and commercialise cilta-cel.7

In April 2021, Janssen announced its submission of a Marketing Authorisation Application (MAA) to the European Medicines Agency seeking approval of cilta-cel for the treatment of patients with relapsed and/or refractory multiple myeloma.8 In December 2020, Janssen announced initiation of a rolling submission of its Biologics License Application (BLA) to the United States (U.S.) Food and Drug Administration (FDA) for cilta-cel.9 In addition to U.S. Breakthrough Therapy Designation in December 2019, cilta-cel received a PRIority MEdicines (PRiME) designation from the European Commission in April 2019, and a Breakthrough Therapy Designation in China in August 2020.10,11 Janssen also received Orphan Drug Designation for cilta-cel from the European Commission in February 2020.12

About Multiple Myeloma
Multiple myeloma is currently an incurable blood cancer that affects a type of white blood cell called plasma cells, which are found in the bone marrow.13 When damaged, these plasma cells rapidly spread and replace normal cells with tumors in the bone marrow.13 In Europe, more than 50,900 people were diagnosed with multiple myeloma in 2020, and more than 32,500 patients died.14 While some patients with multiple myeloma have no symptoms, most patients are diagnosed due to symptoms, which can include bone fracture or pain, low red blood cell counts, tiredness, high calcium levels, kidney problems or infections.15