Ratio Therapeutics Launches to Discover and Drive Early Clinical Development of Best-in-Class Targeted Radiopharmaceuticals for Treatment of Cancers

On June 10, 2022 Ratio Therapeutics Inc. reported its launch with a mission to develop best-in-class targeted radiopharmaceuticals for the treatment of cancers (Press release, Ratio Therapeutics, JUN 10, 2022, View Source [SID1234615894]). Founded by entrepreneurial scientists Jack Hoppin, Ph.D., and John Babich, Ph.D., Ratio emerges from stealth mode with more than $20 million in seed funding, fully funded development alliances with Bayer AG and Lantheus Holdings Inc., a robust portfolio of assets developed with two proprietary technologies, and a growing team of world-class experts in radiopharmaceuticals discovery and development. The company’s near-term plans call for the submission of its first investigational new drug (IND) applications, which are expected this quarter, and the initiation of clinical trials later this year. Based in Boston, Ratio is set to move to a new 19,000-square-foot headquarters and research facility in the Seaport District in January.

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!

Ratio’s radiopharmaceuticals strategy is focused on optimizing tumor localization while minimizing uptake by normal tissues. To achieve this, the company uses its proprietary technology platform called Trillium that is based on Dr. Babich’s prior research at Weill-Cornell Medical College and first developed and validated by the founders in a previous venture. Trillium is a trifunctional small molecule scaffold that can be fine-tuned to alter its plasma clearance, target affinity and therapeutic payload. Each component of the scaffolding can be independently optimized to boost tumor uptake over normal tissue uptake, thereby maximizing therapeutic index. Ratio has successfully applied this framework to several tumor targets and multiple therapeutic payloads.

In addition, Ratio is developing a technology platform to take advantage of the tumor killing power of the alpha emitter, Actinium-225. This proprietary technology is called the Macropa chelate platform. Ratio’s scientists have already successfully incorporated Macropa into the Trillium platform as well as several peptides and antibodies. Macropa’s unique chemistry enables ease of manufacture and robust in vitro and in vivo stability of the resulting radiotherapeutic compound.

"The ability to fine-tune our targeted radiotherapeutics using Trillium and Macropa enables us to address head-on the trifecta of typical challenges we see with most radiopharmaceuticals: delivery, safety and efficacy," said Dr. Babich, Ratio’s President and Chief Scientific Officer. "Over the past year, we have generated significant preclinical data that demonstrate our ability to create excellent performing drug candidates that now are advancing into the clinic. Our goal is to become the partner of choice for pharmaceutical companies committed to this area of cancer therapy by enabling the optimization of a broad array of targeting compounds. We will shepherd these therapies through early clinical studies on our own or in collaboration."

Dr. Hoppin, Ratio’s Chairman and Chief Executive Officer, added, "Targeted radiotherapy is an exciting and emerging field where chemistry meets physics meets medicine. We have assembled and will continue to build a world-class interdisciplinary team of researchers and developers with a singular focus on delivering these treatments to cancer patients. It isn’t often that a start-up company has in place the early financial backing and industry support to advance entirely new drug discoveries to clinical development at this pace. It is with great pride that we announce our formal launch and exit from stealth mode."

In collaboration with Bayer, Ratio has leveraged its Trillium platform for the identification of lead prostate-specific membrane antigen (PSMA)-targeted therapeutic compounds for prostate cancer. At the same time, Ratio is working with Lantheus to develop a lead fibroblast activation protein (FAP)-targeted PET diagnostic compound for a broad array of epithelial-derived cancers, such as breast, pancreatic, lung and stomach cancer. Both collaborations are fully funded and reflect the types of partnerships that Ratio is currently pursuing with other companies.

Disc Medicine Presents Positive Results from Phase 1 Clinical Study of DISC-0974 in Healthy Volunteers at the 2022 EHA Annual Congress

On June 10, 2022 Disc Medicine, a biotechnology company dedicated to the discovery and development of novel therapeutic candidates for serious and debilitating hematologic diseases, reported the results from its phase 1 clinical study of DISC-0974, a first-in-class, monoclonal antibody designed to suppress hepcidin production by inhibiting hemojuvelin (HJV), at the European Hematology Association (EHA) (Free EHA Whitepaper) Congress being held in Vienna, Austria (Press release, Disc Medicine, JUN 10, 2022, View Source [SID1234615893]). The study data demonstrated robust increases in serum iron and markers of erythropoiesis, decreases in hepcidin levels, and that DISC-0974 was well-tolerated in healthy volunteers, consistent with the mechanism of action.

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 very excited by the robust activity and encouraging initial safety profile of DISC-0974 observed in this study and which provide strong validation for our approach to targeting the hepcidin axis," said John Quisel, J.D., Ph.D., Chief Executive Officer. "Having established clinical proof-of-mechanism, we intend to develop DISC-0974 across a wide range of anemias caused by elevated hepcidin, beginning with studies in patients with myelofibrosis and in patients with non-dialysis dependent chronic kidney disease, both of which we plan to initiate this year."

The phase 1 study was a double-blind, placebo-controlled, single-ascending trial in healthy volunteers to evaluate the safety, tolerability, pharmacokinetics and pharmacodynamics of DISC-0974. In the study, 34 healthy, adult subjects received either a single dose of DISC-0974 at escalating dose levels (7 mg IV, 14 mg SC, 28 mg SC, or 56 mg SC) or placebo. The study included endpoints to assess pharmacodynamic activity including serum hepcidin, serum iron and transferring saturation, ferritin levels, and measures of erythropoiesis.

Key data presented:

Rapid, dose-dependent and sustained decrease in serum hepcidin and a corresponding, robust increase in measures of circulating iron, including more than a doubling of transferrin saturation from baseline at the highest dose level

Changes in serum iron also corresponded with markers of iron mobilization and erythropoiesis, including decreased ferritin levels, increased reticulocyte hemoglobin, and increased mean corpuscular hemoglobin

At the 56 mg SC dose level, a single administration of DISC-0974 resulted in a statistically significant improvement in hemoglobin compared to placebo (+1.1 g/dL, p=0.009) at Day 42 and a marked increase in red blood cell count

DISC-0974 was well-tolerated at all dose levels with no serious or severe adverse events, no adverse events leading to study withdrawal, and no adverse event greater than Grade 1

Plasma exposure was dose-related in the 14 to 56 mg SC range and effects were observed through 28 days post-dose, indicating a sustained and clinically meaningful duration of action
"Hepcidin has long been known as the master regulatory hormone of systemic iron homeostasis. However, despite its clear biological importance and role in driving anemia across a host of diseases, no agents that primarily target excess hepcidin have been successfully developed," said Elizabeta Nemeth, Ph.D., Professor of Medicine at the David Geffen School of Medicine at UCLA, and Director of the UCLA Center for Iron Disorders. "The dramatic effects of DISC-0974 on iron and erythropoiesis in this study of healthy volunteers are encouraging and I look forward to following its progress in future clinical trials."

These data were presented at the European Hematology Association (EHA) (Free EHA Whitepaper) Annual Congress in Vienna and the poster is available on the EHA (Free EHA Whitepaper) Congress platform at www.ehaweb.org.

About DISC-0974

DISC-0974 is an investigational, first-in-class monoclonal antibody designed to suppress hepcidin production by inhibiting the hemojuvelin (HJV) co-receptor, a highly selective and critical target of the hepcidin pathway with biological activity that has been validated by human genetic evidence. Hepcidin is the primary regulatory hormone of iron homeostasis and plays a central role by restricting iron absorption and preventing deployment from internal iron stores. DISC-0974 is currently being studied in a phase 1 clinical study of healthy volunteers and is being developed as a potential treatment for anemia of inflammation by suppressing hepcidin and enhancing iron availability for erythropoiesis.

DISC-0974 is an investigational therapy that is not approved for any use in any country. Disc obtained global rights to DISC-0974 and related molecules under a license agreement from AbbVie in October 2019.

Keymed Biosciences Announces Dosing of First Patient in Phase I Trial of Bispecific Antibody CM350

On June 10, 2022 Keymed Biosciences (HKEX: 02162) reported that the first patient has been dosed in the Phase I trial of CM350. CM350 is a GPC3xCD3 bispecific antibody developed by the Company for the treatment of solid tumors (Press release, Keymed Biosciences, JUN 10, 2022, View Source [SID1234615892]). The phase I trial is being conducted to evaluate the safety, tolerability, pharmacokinetics, pharmacodynamics and preliminary antitumor activity of CM350 in patients with 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!

About CM350

Developed on Keymed’s proprietary Novel T cell engager (nTCE) bispecific antibody platform, CM350 is the first GPC3xCD3 bispecific antibody to enter clinical development in China and the second in the world. GPC3 expression is rarely found in normal human adult tissues, but highly upregulated in a variety of solid tumors including hepatocellular carcinoma, lung cancer, gastric cancer and esophageal cancer, suggesting that GPC3 is an ideal target for the therapeutics of multiple solid tumors, especially hepatocellular carcinoma.

Preclinical studies have shown that CM350 could effectively kill GPC3-positive tumor cells by T cell-dependent cellular cytotoxicity (TDCC) and exert highly potent antitumor activity in mouse tumor models. Based on the advantages of the nTCE bispecific antibody platform, CM350 is optimized with highly effective tumor cell killing with favorable safety profile by minimizing non-specific effect on normal cells and cytokine release. Therefore, CM350 may provide a promising treatment option for cancer patients with better efficacy and manageable safety.

About nTCE bispecific antibody platform

Novel T cell engager (nTCE) is a CD3 bispecific antibody technology platform developed by Keymed with proprietary rights. Antibodies developed by the nTCE platform can effectively kill tumor cells by TDCC with minimized non-specific effect on normal cells and cytokine release, thereby reducing the occurrence of potential cytokine release syndrome in clinical treatment.

Antibodies developed by our nTCE platform have similar structures to native antibodies with favorite pharmacokinetic profiles. Through the sophisticated protein engineering technology, nTCE platform enables efficient pairing of cognate heavy/light chains of the bispecific antibody with high yield and low aggregation. Using our advanced nTCE platform, Keymed has developed a series of bispecific antibodies currently in the stages of preclinical and clinical development.

Ginkgo Bioworks Announces Participation in Goldman Sachs 43rd Annual Global Healthcare Conference

On June 10, 2022 Ginkgo Bioworks (NYSE: DNA), the leading horizontal platform for cell programming, reported that management is scheduled to participate in Goldman Sachs’ 43rd Annual Global Healthcare Conference, on June 15, 2022, at 10:40 a.m. PT (Press release, Ginkgo Bioworks, JUN 10, 2022, View Source [SID1234615891]).

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!

Further details, a webcast link, and a replay of the presentation, if available, will be posted on the company’s investor relations website at View Source

Janssen Presents Updated Data at EHA for Teclistamab in Patients with Relapsed or Refractory Multiple Myeloma

On June 10, 2022 The Janssen Pharmaceutical Companies of Johnson & Johnson reported that updated efficacy and safety results from the teclistamab cohort of the Phase 1b TriMM-2 study (NCT04108195) (Press release, Johnson & Johnson, JUN 10, 2022, View Source [SID1234615890]).1 Teclistamab, an investigational, off-the-shelf, T-cell redirecting bispecific antibody targeting B-cell maturation antigen (BCMA) and CD3 on T-cells, is being studied in combination with the anti-CD38 monoclonal antibody, DARZALEX (daratumumab) subcutaneous (SC) formulation, in patients with relapsed or refractory multiple myeloma (RRMM) who have received three or more prior lines of therapy.1 Patients in the study, including a high proportion with prior anti-CD38 monoclonal antibody exposure, achieved encouraging overall response rates (ORR) with this combination treatment.1 These data will be presented at the 2022 European Hematology Association (EHA) (Free EHA Whitepaper) Annual Congress as an oral presentation on Sunday, 12 June (Abstract S188).

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!

At a median follow-up of 8.6 months (range, 0.3-19.6), 76.5 percent (39/51) of response-evaluable patients enrolled in the study achieved a response, including 36 patients (70.6 percent) who achieved a very good partial response (VGPR) or better.1 In patients with prior anti-CD38 exposure, an ORR of 73.7 percent was achieved.1 The median time to first confirmed response was one month, and responses remained durable and deepened over time.1 At the analysis cut-off, 66.7 percent of patients who achieved a response (26/39) were alive and continuing on therapy.1

"Responders to the combination of teclistamab plus subcutaneous daratumumab included patients with prior exposure to BCMA or anti-CD38 targeted agents, which is encouraging," said Paula Rodríguez-Otero†, M.D., Ph.D., Department of Hematology, Clínica Universidad de Navarra, Pamplona, Spain and principal study investigator. "These data also suggest this steroid-sparing regimen may lead to a clinically efficacious regimen in highly refractory patients."

The open-label, multicentre, multicohort Phase 1b TriMM-2 study is investigating the safety and efficacy of teclistamab in combination with daratumumab SC for patients with RRMM.1 Enrolled patients received a median of five prior lines of therapy, 58.5 percent were triple-class refractory, 30.8 percent were penta-drug refractory, and 63.1 percent were refractory to anti-CD38 treatment.1 Eighty percent of patients were refractory to their last line of therapy.1

As of April 6, 2022, 65 patients received daratumumab SC 1,800mg at the approved schedule plus teclistamab 1.5mg/kg weekly (QW) or 3mg/kg every other week (Q2W) subcutaneously.1 Pre-medications, including steroids, were limited to the two step-up doses and the first full dose of teclistamab.1 Treatment with the combination regimen were tolerable and no unexpected or overlapping toxicities were observed.1 The most common adverse events (AEs) were cytokine release syndrome (CRS) (67.7 percent, all Grade 1 or 2); neutropenia (49.2 percent, 41.5 percent Grade 3 or 4); and anaemia (41.5 percent, 27.7 percent Grade 3 or 4).1 One patient (two percent) had Grade 1 immune effector cell–associated neurotoxicity syndrome (ICANS) which fully resolved.1 Infections were experienced by 67.7 percent of patients (27.7 percent Grade 3 or 4).1 Four patients died from AEs, all unrelated to teclistamab or daratumumab SC treatment.1

"For nearly 20 years, we have been committed to overcoming multiple myeloma and an important part of our strategy is to continue to invest and develop complementary and combinable regimens that improve outcomes for patients and their caregivers," said Edmond Chan MBChB M.D. (Res), EMEA Therapeutic Area Lead Haematology, Janssen-Cilag Limited. "Daratumumab has become a foundational therapy for multiple myeloma, and it is exciting to see how we can continue to grow its potential through combinations with novel treatments."

Pharmacodynamic analyses demonstrate that the combination upregulates CD38+/CD8+ T-cells and proinflammatory cytokines, suggesting the potential for complementary activity.1 Additional studies are needed to fully understand the potential clinical benefit of this biological activity.

The efficacy and pharmacodynamic profile of teclistamab in combination with daratumumab SC in patients refractory to anti-CD38 therapy suggest that higher response rates may be observed in patients with anti-CD38 naïve or sensitive disease who are enrolling in the MajesTEC-3 study (NCT05083169).1,2 The ongoing Phase 3 MajesTEC-3 study compares the efficacy of the teclistamab-daratumumab combination with daratumumab SC in combination with pomalidomide and dexamethasone (DPd) or daratumumab SC in combination with bortezomib and dexamethasone (DVd).2 Patients in the trial must have received one to three prior lines of therapy including a proteasome inhibitor (PI) and lenalidomide; patients who have received only one prior line of therapy must be refractory to lenalidomide.2 Patients who have progressed on or within 60 days of the last dose of lenalidomide given as maintenance therapy are also included.2

"These data suggest the potential of a fully immune-based regimen for patients with heavily pretreated multiple myeloma," said Yusri Elsayed, M.D., M.HSc., Ph.D., Vice President, Disease Area Leader, Hematologic Malignancies, Janssen Research & Development, LLC. "We are committed to the ongoing development of this combination and other treatments for patients who remain in need of new options."

#ENDS#

About Teclistamab
Teclistamab is an investigational, fully humanised, T-cell redirecting, IgG4 bispecific antibody targeting both BCMA (B-cell maturation antigen) and CD3, on T-cells.1 BCMA is expressed at high levels on multiple myeloma cells.3,4,5,6,7 Teclistamab redirects CD3-positive T-cells to BCMA-expressing myeloma cells to induce killing of tumor cells.8

Teclistamab is currently being evaluated in several monotherapy and combination studies.9,10,11,12,13 In 2020, the European Commission (EC) and the United Stated (U.S.) Food and Drug Administration (FDA) both granted teclistamab Orphan Drug Designation for the treatment of multiple myeloma. In January 2021 and June 2021, teclistamab received a PRIority MEdicines (PRIME) designation by the European Medicines Agency (EMA) and Breakthrough Therapy Designation (BTD) by the U.S. FDA, respectively. PRIME offers enhanced interaction and early dialogue to optimise drug development plans and speed up evaluation of cutting-edge, scientific advances that target a high unmet medical need.14 The FDA grants BTD to expedite the development and regulatory review of an investigational medicine that is intended to treat a serious or life-threatening condition based on preliminary clinical evidence that demonstrates the drug may have substantial improvement in at least one clinically significant endpoint over available therapy.15 In December 2021, Janssen submitted a Biologics License Application (BLA) to the FDA seeking approval of teclistamab for the treatment of patients with RRMM; a marketing authorisation application (MAA) was submitted to the EMA for teclistamab approval in January 2022.

About daratumumab and daratumumab SC
Janssen is committed to exploring the potential of daratumumab for patients with multiple myeloma across the spectrum of the disease. Daratumumab has been approved in eight indications for multiple myeloma, three of which are in the frontline setting, including newly diagnosed patients who are transplant eligible and ineligible.16

In August 2012, Janssen Biotech, Inc. and Genmab A/S entered a worldwide agreement, which granted Janssen an exclusive license to develop, manufacture and commercialise daratumumab. Since launch, daratumumab has become a foundational therapy in the treatment of multiple myeloma, having been used in the treatment of more than 274,000 patients worldwide.17 Daratumumab is the only CD38-directed antibody approved to be given subcutaneously to treat patients with multiple myeloma. Daratumumab SC is co-formulated with recombinant human hyaluronidase PH20 (rHuPH20), Halozyme’s ENHANZE drug delivery technology.18

CD38 is a surface protein that is present in high numbers on multiple myeloma cells, regardless of the stage of disease.16 Daratumumab binds to CD38 and inhibits tumour cell growth causing myeloma cell death.16 Daratumumab may also have an effect on normal cells.16 Data across eight Phase 3 clinical trials, in both the frontline and relapsed settings, have shown that daratumumab based regimens resulted in significant improvement in PFS and/or OS.19,20,21,22,23,24,25,26

For further information on daratumumab, please see the Summary of Product Characteristics at: View Source

About Multiple Myeloma
Multiple myeloma is an incurable blood cancer that affects a type of white blood cell called plasma cells, which are found in the bone marrow.27 In multiple myeloma, cancerous plasma cells change and grow out of control.27 In Europe, more than 50,900 people were diagnosed with multiple myeloma in 2020, and more than 32,500 patients died.28 While some patients with multiple myeloma initially 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 or kidney failure.29