Akeso Presented Promising Results of Cadonilimab and Lenvatinib in Combination with TACE in uHCC at 2024 ASCO GI

On January 23, 2024 Akeso, Inc. (9926.HK) ("Akeso," "we," or the "Company") reported that the Company presented the promising phase II results of cadonilimab (a tetravalent PD-1/CTLA-4 bispecific antibody) and Lenvatinib (Len) combined with TACE for the treatment of mid- and advanced-stage hepatocellular carcinoma (HCC) in a poster at the 2024 American Society of Clinical Oncology (ASCO) (Free ASCO Whitepaper) Gastrointestinal Cancers Symposium in San Francisco, California (Press release, Akeso Biopharma, JAN 23, 2024, View Source [SID1234639437]). Based on the encouraging results, the company plans to further explore this combination therapy for the mid- and advanced-stage HCC indication.

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!

AK104-216 is an open-label, multicenter phase II trial (NCT05319431). As of August 15, 2023, 60 patients received at least once Cadonilimab and the median duration of follow-up was 5.1 months. Cadonilimab and Lenvatinib combined with TACE showed great tumor response: The ORR and DCR were 85.0% and 95% per mRECIST. 15% subjects achieved CR and 70% subjects achieved PR. The median PFS was not reached, 6-month PFS rate and 9-month PFS rate was 75.6% and 60.4%, respectively. Cadonilimab and Lenvatinib combined with TACE showed manageable safety in mid- and advanced-stage HCC.

HCC typically has an insidious onset, with over 80% of patients being diagnosed in the middle to late stages of the disease. The low rate of radical resection and poor overall prognosis further compound the challenges associated with HCC. Transarterial chemoembolization (TACE) is a standard therapy recommended for intermediate-stage unresectable hepatocellular carcinoma (uHCC) but has unsatisfying tumor control due to the increasing risk of tumor angiogenesis. Immune-checkpiont inhibitors, in combination with targeted therapy and local therapy, have been developed as promising treatment for mid- and advanced-stage HCC.

Cadonilimab and Lenvatinib combined with TACE have demonstrated promising efficacy and manageable toxicity in treating mid- and advanced-stage HCC. Patients can expect to benefit from tumor progression control after TACE and lasting survival. Akeso is actively preparing a clinical study of this therapy in mid- and advanced-stage HCC, which will be registrational. Furthermore, Akeso is accelerating the progress of the registrational Phase III clinical study of cadonilimab monotherapy for postoperative adjuvant treatment of hepatocellular carcinoma. The enrollment for this study is expected to be completed in 2024.

Taiho Oncology Announces Publication of Final Results of the Phase 3 ASCERTAIN Clinical Trial of Oral Decitabine and Cedazuridine Fixed Dose Combination (INQOVI®) in Patients With MDS and CMML

On January 23, 2024 Taiho Oncology, Inc. reported publication of the final results from the pivotal ASCERTAIN clinical trial of fixed-dose oral decitabine and cedazuridine (INQOVI) compared to intravenous decitabine in adults with intermediate and high-risk myelodysplastic syndromes (MDS) including chronic myelomonocytic leukemia (CMML) (Press release, Taiho, JAN 23, 2024, View Source [SID1234639436]).

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 ASCERTAIN trial was the first Phase 3 trial to demonstrate pharmacologic equivalence between an oral and an intravenous (IV) formulation of a hypomethylating agent for use in the treatment of patients with MDS or CMML. As reported in the January 2 issue of The Lancet Haematology, median overall survival (mOS) in the trial population was approximately 32 months.1 In addition, the overall response rate was 62% in the intent to treat patient population. The percentage of patients in this trial who moved to transplantation reached 20%, exceeding expected transplantation rates in patients receiving hypomethylating agents for MDS and CMML.1

Safety findings from the study were comparable with those previously observed with IV decitabine. The most common treatment-emergent adverse events of thrombocytopenia, neutropenia and anemia were consistent with expected adverse events with parenteral hypomethylating agent treatment.

The data from the study supported the simultaneous approval of INQOVI by the U.S. Food and Drug Administration and Health Canada in July 2020 for the treatment of adult patients with myelodysplastic syndromes (MDS), including previously treated and untreated, de novo and secondary MDS with the following French-American-British subtypes (refractory anemia, refractory anemia with ringed sideroblasts, refractory anemia with excess blasts, and chronic myelomonocytic leukemia [CMML]) and intermediate-1, intermediate-2, and high-risk International Prognostic Scoring System groups.2

"Until recently, azacitidine and decitabine, both widely used hypomethylating agents, were available only in parenteral form, requiring patients with MDS and CMML to travel to treatment centers daily for 5 or 7 consecutive days of each 28-day treatment cycle," said Guillermo Garcia-Manero, MD, Professor, Department of Leukemia, Division of Cancer Medicine, the University of Texas MD Anderson Cancer Center, Houston, and the lead author on the publication. "The ASCERTAIN study has demonstrated that the orally delivered fixed dose combination of decitabine and cedazuridine is an alternative option to parenteral administration of decitabine for patients with these diseases. The observed median overall survival of greater than 30 months in the ASCERTAIN study compared with historical controls is encouraging."

Added Tehseen Salimi, MD, MHA, Senior Vice President and Head of Medical Affairs, Taiho Oncology, Inc., "Patients living with MDS and CMML can benefit from the convenience of an at-home hypomethylating agent treatment that may potentially reduce the number of office visits and the travel that comes with it."

About the ASCERTAIN Trial
The Phase 3 ASCERTAIN clinical trial was a multicenter, randomized, open-label, crossover pharmacokinetics (PK) study comparing oral decitabine (35mg) and cedazuridine (100mg) fixed-dose combination tablet given once daily for 5 days on a 28-day cycle to IV decitabine (20mg/m2) administered as a daily 1-hour IV infusion for 5 days on a 28-day cycle, in the first 2 cycles in patients with MDS and CMML. Patients continued to receive oral decitabine and cedazuridine from Cycle 3 onwards. The primary endpoint of the study was total 5-day area-under-the-curve (AUC) equivalence of oral decitabine and cedazuridine and IV decitabine.

For more information about ASCERTAIN, please visit: View Source;draw=3&rank=19.

INDICATIONS
Decitabine and cedazuridine, marketed under the brand name INQOVI, is indicated for treatment of adult patients with myelodysplastic syndromes (MDS), including previously treated and untreated, de novo and secondary MDS with the following French-American-British subtypes (refractory anemia, refractory anemia with ringed sideroblasts, refractory anemia with excess blasts, and chronic myelomonocytic leukemia [CMML]) and intermediate-1, intermediate-2, and high-risk International Prognostic Scoring System groups.2

INQOVI is the first and only oral hypomethylating agent approved by the FDA and by Health Canada for the treatment of adults with intermediate and high-risk MDS including CMML.

Commercialization of INQOVI in the U.S. and Canada is conducted by Taiho Oncology, Inc. and Taiho Pharma Canada, Inc., respectively.

IMPORTANT SAFETY INFORMATION WARNINGS AND PRECAUTIONS

Myelosuppression
Fatal and serious myelosuppression can occur with INQOVI. Based on laboratory values, new or worsening thrombocytopenia occurred in 82% of patients, with Grade 3 or 4 occurring in 76%. Neutropenia occurred in 73% of patients, with Grade 3 or 4 occurring in 71%. Anemia occurred in 71% of patients, with Grade 3 or 4 occurring in 55%. Febrile neutropenia occurred in 33% of patients, with Grade 3 or 4 occurring in 32%. Myelosuppression (thrombocytopenia, neutropenia, anemia, and febrile neutropenia) is the most frequent cause of INQOVI dose reduction or interruption, occurring in 36% of patients. Permanent discontinuation due to myelosuppression (febrile neutropenia) occurred in 1% of patients. Myelosuppression and worsening neutropenia may occur more frequently in the first or second treatment cycles and may not necessarily indicate progression of underlying MDS.

Fatal and serious infectious complications can occur with INQOVI. Pneumonia occurred in 21% of patients, with Grade 3 or 4 occurring in 15%. Sepsis occurred in 14% of patients, with Grade 3 or 4 occurring in 11%. Fatal pneumonia occurred in 1% of patients, fatal sepsis in 1%, and fatal septic shock in 1%.

Obtain complete blood cell counts prior to initiation of INQOVI, prior to each cycle, and as clinically indicated to monitor response and toxicity. Administer growth factors and anti‑infective therapies for treatment or prophylaxis as appropriate. Delay the next cycle and resume at the same or reduced dose as recommended.

Embryo-Fetal Toxicity
INQOVI can cause fetal harm. Advise pregnant women of the potential risk to a fetus. Advise patients to use effective contraception during treatment and for 6 months (females) or 3 months (males) after last dose.

ADVERSE REACTIONS
Serious adverse reactions in > 5% of patients included febrile neutropenia (30%), pneumonia (14%), and sepsis (13%). Fatal adverse reactions included sepsis (1%), septic shock (1%), pneumonia (1%), respiratory failure (1%), and one case each of cerebral hemorrhage and sudden death.

The most common adverse reactions (≥ 20%) were fatigue (55%), constipation (44%), hemorrhage (43%), myalgia (42%), mucositis (41%), arthralgia (40%), nausea (40%), dyspnea (38%), diarrhea (37%), rash (33%), dizziness (33%), febrile neutropenia (33%), edema (30%), headache (30%), cough (28%), decreased appetite (24%), upper respiratory tract infection (23%), pneumonia (21%), and transaminase increased (21%). The most common Grade 3 or 4 laboratory abnormalities (≥ 50%) were leukocytes decreased (81%), platelet count decreased (76%), neutrophil count decreased (71%), and hemoglobin decreased (55%).

USE IN SPECIFIC POPULATIONS

Lactation
Because of the potential for serious adverse reactions in the breastfed child, advise women not to breastfeed during treatment with INQOVI and for 2 weeks after the last dose.

Renal Impairment
No dosage modification of INQOVI is recommended for patients with mild or moderate renal impairment (creatinine clearance [CLcr] of 30 to 89 mL/min based on Cockcroft-Gault). Due to the potential for increased adverse reactions, monitor patients with moderate renal impairment (CLcr 30 to 59 mL/min) frequently for adverse reactions. INQOVI has not been studied in patients with severe renal impairment (CLcr 15 to 29 mL/min) or end-stage renal disease (ESRD: CLcr <15 mL/min).

Please see the accompanying Full Prescribing Information.

About Myelodysplastic Syndromes (MDS) and Chronic Myelomonocytic Leukemia (CMML)
Myelodysplastic syndromes are a heterogeneous group of hematopoietic stem cell disorders characterized by dysplastic changes in myeloid, erythroid, and megakaryocytic progenitor cells, and associated with cytopenias affecting one or more of the three lineages. U.S. incidence of MDS is estimated to be 10,000 cases per year, although the condition is thought to be under-diagnosed.3,4 The prevalence has been estimated to be from 60,000 to 170,000 in the U.S.5 MDS may evolve into acute myeloid leukemia (AML) in one-third of patients.6 The prognosis for MDS patients is poor; patients die from complications associated with cytopenias (infections and bleeding) or from transformation to AML.

CMML is a clonal hematopoietic malignancy characterized by accumulation of abnormal monocytes in the bone marrow and in blood. The incidence of CMML in the U.S. is approximately 1,100 new cases per year,7 and CMML may transform into AML in 15% to 30% of patients.8

About Decitabine and Cedazuridine Fixed-Dose Combination
This product is an orally administered, fixed dose combination of the approved anti-cancer DNA hypomethylating agent, decitabine, together with cedazuridine,3 an inhibitor of cytidine deaminase.4 By inhibiting cytidine deaminase in the gut and the liver, the fixed dose combination is designed to allow for oral delivery of decitabine over five days in a given cycle to achieve comparable systemic exposure to IV decitabine administered over five days.

The oral decitabine and cedazuridine fixed-dose combination has been evaluated in a Phase 1/2 pharmacokinetics-guided dose escalation and dose confirmation study, and a Phase 3 exposure equivalence study in patients with MDS and CMML – the ASCERTAIN study.

PharmAbcine Announces First Patient Dosed in Phase 1a/b Clinical Trial of PMC-309 in Combination with KEYTRUDA® (pembrolizumab) in Patients with Advanced or Metastatic Solid Tumors

On January 23, 2024 PharmAbcine Inc. (KOSDAQ: 208340ks), a clinical-stage biotech company focusing on next-generation antibody therapeutics, reported the initiation of patient dosing in the Phase 1a/b clinical trial of PMC-309 in patients with advanced or metastatic solid tumors (Press release, PharmAbcine, JAN 23, 2024, View Source [SID1234639435]).

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!

PMC-309 is an IgG1 monoclonal antibody with specific binding to VISTA in immunosuppressive cells, exhibiting excellent binding affinity at various pH conditions within the tumor microenvironment (TME). By inhibiting VISTA, PMC-309 presents a differentiated mechanism of action contributing to anti-cancer effects through activation of T cells, activation of monocytes, and proliferation of M1 macrophages.

This open-label clinical trial comprises two phases, Phase 1a and Phase 1b. Phase 1a involves PMC-309 monotherapy and combination therapy with KEYTRUDA (pembrolizumab), determining the maximum tolerated dose (MTD) and the recommended Phase 2 dose (RP2D). In Phase 1b, safety and tolerability of PMC-309 monotherapy and combination therapy with KEYTRUDA at the RP2D will be evaluated. The clinical trial, spanning four institutions in Australia, aims to enroll a total of 67 patients.

Dr. Jin-San Yoo, President and Chief Executive Officer of PharmAbcine, stated, "This clinical trial aims to assess the human safety of PMC-309 and explore the potential of both monotherapy and combination therapy with KEYTRUDA. We are committed to developing new therapeutic options for cancer patients facing high unmet needs, emphasizing our dedication to advancing oncology treatments."

For more information on the clinical trial, please visit clinicaltrials.gov, identifier NCT 05957081.

KEYTRUDA is a registered trademark of Merck Sharp & Dohme LLC, a subsidiary of Merck & Co., Inc., Rahway, NJ, USA.

Harbour BioMed Announces IND Clearance for HBM9027 in the U.S.

On January 23, 2024 Harbour BioMed (the "Company", HKEX: 02142) reported that the Company has been granted the clearance of Investigational New Drug (IND) from the Food and Drug Administration (FDA) of the United States to initiate the first-in-human (FIH) clinical trial in the U.S. for bispecific antibody HBM9027 (Press release, Harbour BioMed, JAN 23, 2024, View Source [SID1234639434]). This is a phase I study to evaluate the safety, tolerability, pharmacokinetics, and anti-tumor activity of HBM9027 in subjects with advanced solid tumors.

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!

HBM9027 is generated from Harbour BioMed’s proprietary fully human HBICE platform. It is a novel PD-L1xCD40 bispecific antibody, which is designed to activate CD40 relied on PD-L1 crosslinking for a promising safety profile. PD-L1 is overexpressed on a variety of solid malignancies, and with its crosslinking dependent specificity on tumors and potent immune modulation activity, HBM9027 has shown excellent and promising safety profile with strong anti-tumor efficacy in its pre-clinical studies.

"HBM9027, developed through our proprietary HBICE platform, has demonstrated a promising safety profile and robust anti-tumor efficacy," said Dr. Jingsong Wang, Founder, Chairman and CEO of Harbour BioMed. "This achievement highlights the capability of our technology platform in developing novel solutions for advanced solid tumors. We firmly believe that our unweaving commitment to innovation will bring about meaningful advancements in cancer treatment."

About HBM9027

HBM9027 is a novel PD-L1xCD40 bispecific antibody, developed using the HBICE bispecific technology and Harbour Mice Platform. The development of PD-L1xCD40 bispecific HBICE further expands the Company’s bispecific immune cell engager into the cutting-edge DC/myeloid cell engager field and demonstrates HBICE platform’s versatile geometry formats and plug-and-play advantages.

Papyrus Therapeutics Receives Approval of New Patent Claims for Multi-Cancer Tumor Suppressor Therapy Platform

On January 23, 2024 Papyrus Therapeutics Inc., a biotechnology platform company for the development of potent, multi-cancer, tumor suppressor precision therapies for solid cancers, reported that the United States Patent and Trademark Office (USPTO) granted approval of all 20 claims in Papyrus’ submitted Patent (Press release, Papyrus Therapeutics, JAN 23, 2024, View Source;utm_medium=rss&utm_campaign=papyrus-therapeutics-receives-approval-of-new-patent-claims-for-multi-cancer-tumor-suppressor-therapy-platform [SID1234639433]). That the USPTO approved all 20 claims is distinguishing given the infrequency patent grants are awarded without prior art being cited against the claims.

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 nature of the expanded patent recognizes the groundbreaking novelty and potential of Papyrus’ platform’s science, knowledgebase, and AI enablement to streamline the discovery of naturally occurring tumor suppressor proteins like OPCML as therapeutics. Such proteins, when identified as silenced in human cancer, can be restored in those cancers by Papyrus’ recombinant-OPCML (r-OPCML) protein replacement therapy through intravenous delivery. Unlike chemotherapy and other oncogene-focused therapeutics, r-OPCML targets only cancer cells deficient in this protein without damaging healthy cells that normally express it. Further, the data-to-date points to the therapeutic being a well-tolerated and effective treatment.

According to Papyrus’ co-founder / chief scientific officer, and renowned translational medical oncologist Hani Gabra, PhD FRCP, "Preclinical studies of Papyrus’ lead clinical asset PYTX-004, the r-OPCML-Fc fusion tumor suppressor analog, have demonstrated a high degree of tumor suppressor efficacy as a monotherapy and as a combination therapy in ovarian cancer with no adverse toxicities noted thus far. This, in concert with USPTO’s approval of all 20 claims reinforces the transformational potential of the Papyrus tumor suppressor discovery platform to enable a pipeline of novel, cancer-targeted protein-replacement tumor suppressor therapeutics." Similar results for this suppressor replacement therapy have been observed for HER2+ breast cancer and are projected for other cancers, with loss of this tumor suppressor noted in at least half of all human solid cancers.

OPCML, a member of the IgLON family of proteins, is a naturally occurring tumor suppressor protein. PYTX-004 is Papyrus’ r-OPCML-Fc lead tumor suppressor biotherapeutic in development that is outlined in the patent application. The Company expects to have continued success with upcoming patent filings based on unique synthetic alterations of OPCML and other related IgLON family members that will enable the development of a pipeline of pioneering unique synthetic tumor suppressor biotherapeutics with novel specificities for monotherapy and combination therapies in differing indications.