Janssen Presents Longer-Term Talquetamab Follow-Up Data Showing Overall Response Rates of More Than 70 Percent in Heavily Pretreated Patients with Multiple Myeloma

On June 3, 2023 The Janssen Pharmaceutical Companies of Johnson & Johnson reported updated results from the pivotal Phase 1/2 MonumenTAL-1 study of the investigational bispecific antibody talquetamab in the treatment of patients with relapsed or refractory multiple myeloma (RRMM) (Press release, Johnson & Johnson, JUN 3, 2023, View Source [SID1234632434]). Data from the MonumenTAL-1 study highlight safety and efficacy results (Abstract #8036) and an analysis of infections and parameters of humoral immunity in patients with RRMM treated with talquetamab (Abstract #8020).1,2 These data will be presented at the 2023 American Society of Clinical Oncology (ASCO) (Free ASCO Whitepaper) Annual Meeting.1,2 Additional data from the Phase 2 TRiMM-2 study, evaluating talquetamab in combination with DARZALEX FASPRO (daratumumab and hyaluronidase-fihj), were presented (Abstract #8003) at the meeting.3

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

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

                  Schedule Your 30 min Free Demo!

Patients in the Phase 1/2 MonumenTAL-1 study (n=339) were treated with subcutaneous (SC) talquetamab at the recommended Phase 2 dose (RP2D) of 0.8 mg/kg biweekly (Q2W) or 0.4 mg/kg weekly (QW) with step-up doses.1 The overall response rate (ORR) to talquetamab was similar across both doses.1 With a median follow-up of 12.7 months, 71.7 percent (104/145) of response-evaluable patients treated with the 0.8 mg/kg Q2W dose achieved a response, 60.7 percent achieved a very good partial response (VGPR) or better, nine percent achieved a complete response (CR), and 29.7 percent achieved a stringent complete response.1 With a median follow-up of 18.8 months, 74.1 percent (106/143) of response-evaluable patients treated with the 0.4 mg/kg QW dose achieved a response, 59.4 percent achieved a VGPR or better, 9.8 percent achieved a CR, and 23.8 percent achieved a stringent CR.1 In a separate cohort of patients treated with prior T-cell redirection therapy, 64.7 percent (33/51) achieved a response, and 54.9 percent achieved a VGPR or better, with a median follow-up of 14.8 months.1

"The updated results from the MonumenTAL-1 study continue to show the encouraging potential of talquetamab for heavily pretreated patients with multiple myeloma, including those who may have been exposed to prior T-cell redirection therapy," said Carolina Schinke, M.D., Associate Professor of Medicine at the Myeloma Center at the University of Arkansas for Medical Sciences and principal investigator.† "With a high overall response rate among relapsed or refractory patients, the results underscore the efficacy of talquetamab as a novel option for later-line patients who otherwise face a poor prognosis, including patients with high-risk disease."

Responses were durable; median duration of response (DOR) was not reached for patients on the Q2W dose and was 9.5 months (range, 6.7-13.3) for patients who received QW dosing.1 The 12-month overall survival (OS) rates were 77.4 percent, 76.4 percent and 62.9 percent in the 0.8 mg/kg Q2W dose, 0.4 mg/kg QW dose, and prior T-cell redirection cohorts, respectively.1 The 12-month progression free survival (PFS) rates were 54.4 percent, 34.9 percent and 38.1 percent in the 0.8 mg/kg Q2W dose, 0.4 mg/kg QW dose, and prior T-cell redirection cohorts, respectively.1

Study results showed a low discontinuation rate due to adverse events (AEs) (0.8 mg/kg Q2W dose, eight percent; 0.4 mg/kg QW dose, five percent).1,4 The most common AEs at the 0.8 mg/kg Q2W dose and 0.4 mg/kg QW dose were cytokine release syndrome (CRS; 74.5 percent, 0.7 percent Grade 3/4; 79 percent, 2.1 percent Grade 3/4, respectively); dysgeusia (71 percent and 72 percent, respectively; all Grade 1/2); and skin-related AEs (73.1 percent Grade 1/2, 0.7 percent Grade 3/4; 55.9 percent all Grade 1/2, respectively).1 The safety profile was clinically manageable with low rates of Grade 3 or higher infections (0.8 mg/kg Q2W dose, 14.5 percent; 0.4 mg/kg QW dose, 19.6 percent) and low rates of talquetamab discontinuation due to infection (0.8 mg/kg Q2W dose, zero percent; 0.4 mg/kg QW dose, 1.4 percent).1 Safety in the T-cell redirection subgroup was consistent with what was observed in the weekly and biweekly cohorts.1 New onset infections were primarily limited to the first 100 days.1,2 No new safety signals were observed with longer term follow-up.1,5 There were no talquetamab-related deaths.4

Analysis of Infections and Parameters of Humoral Immunity in Patients with Relapsed/Refractory Multiple Myeloma Treated with Talquetamab Monotherapy in MonumenTAL-1

Patients treated with talquetamab, which targets GPRC5D, an antigen uniquely expressed on plasma cells, showed effective myeloma control with concurrent preservation of humoral immune function (i.e., antibody response by B-cells) and recovery of low blood cell counts, distinguishing talquetamab as an important emerging therapy for RRMM.2 The study results suggest the incidence of infection was less frequent with talquetamab compared with data from studies of B-cell maturation antigen (BCMA)-targeted T-cell–based therapies.2 No decreases in B-cells or polyclonal low serum immunoglobulin G (IgG) were observed, supporting talquetamab as a B-cell–preserving treatment and allowing maintenance of key elements of humoral immunity.2 Of 339 patients, infections occurred in 65.8 percent (20.5 percent Grade 3/4) after median follow-up of 12.7, 18.8, and 14.8 months in the 0.8 mg/kg Q2W, 0.4 mg/kg QW, and prior T-cell redirection cohorts, respectively.2 There were few opportunistic infections, and 1.2 percent of infections led to death.2

Updated data from the Phase 2 TRiMM-2 Study Evaluating Talquetamab in Combination with DARZALEX FASPRO

Results from the Phase 2 TRiMM-2 study showed patients with heavily pretreated multiple myeloma who received the investigational SC combination of talquetamab and DARZALEX FASPRO (daratumumab and hyaluronidase-fihj) achieved deep and durable responses.3 The study included some patients who were previously exposed to anti-CD38, BCMA-targeted, and T-cell redirecting therapies.3

Patients in the TRiMM-2 study were treated with talquetamab at a SC RP2D of 0.8 mg/kg Q2W or 0.4 mg/kg QW (with step-up doses) in addition to DARZALEX FASPRO.3 With a median follow-up of 15 months, 84 percent (42/50) of patients in the 0.8 mg/kg Q2W arm achieved a response, including 74 percent who achieved a VGPR or better, 16 percent who achieved a CR, and 36 percent who achieved a stringent CR.3 The ORR among patients with prior exposure to an anti-CD38 antibody was 82.2 percent (37/45), and 78.9 percent (15/19) of patients with prior treatment with T-cell redirection therapy in the 0.8 mg/kg Q2W cohort responded.3 With a median follow-up of 16.8 months, 71.4 percent (10/14) of patients in the 0.4 mg/kg QW arm achieved a response; 57.1 percent achieved a VGPR or better, 14.3 percent achieved a CR, and 28.6 percent achieved a stringent CR.3 The ORR observed among patients with prior exposure to an anti-CD38 antibody was 63.6 percent (7/11), and the ORR was 66.7 percent (4/6) among patients with prior treatment with T-cell redirection therapy in the 0.4 mg/kg QW cohort.3

At data cutoff, 65.4 percent of responders remained on therapy (63.6 percent and 61.5 percent who were anti-CD38 exposed or refractory, respectively).3 Median DOR was 20.3 months in the 0.8 mg/kg Q2W arm and was not reached in the 0.4 mg/kg QW arm.3 Median PFS was 19.4 months in the 0.8 mg/kg Q2W arm and was not reached in the 0.4 mg/kg QW arm; 12-month mPFS rate was 67.4 percent and 77.4 percent respectively.3 Median OS was not reached in either arm; 12-month OS was 91.5 percent and 92.3 percent in the 0.8 mg/kg Q2W and 0.4 mg/kg QW arms, respectively.3

"The latest results from the TRiMM-2 study further reinforce the potential of talquetamab in combination with subcutaneous daratumumab as an important treatment option for patients, including those previously treated with an anti-CD38 regimen or prior T-cell redirection therapy," said Bhagirathbhai Dholaria, M.D., M.B.B.S., Assistant Professor of Medicine in the Division of Hematology-Oncology at Vanderbilt University Medical Center in Nashville, and principal investigator.† "With an overall response rate of nearly 80 percent, this durable combination provides the potential for significant disease control and survival in heavily pretreated patients with relapsed or refractory multiple myeloma."

The safety profile was clinically manageable with low rates of Grade 3/4 infections (0.8 mg/kg Q2W dose, 25.5 percent; 0.4 mg/kg QW dose, 21.4 percent) and talquetamab discontinuations (1.5 percent).3 Almost all patients (95.4 percent) received antibacterial, antifungal or antiviral prophylaxis. No new safety signals were observed with longer term follow-up.3 The most common non-hematologic AEs at the 0.8 mg/kg Q2W dose and 0.4 mg/kg QW dose cohorts were CRS (80 percent and 71 percent, respectively; all Grade 1/2), skin-related AEs (84 percent and 71 percent, respectively; Grade 3/4: eight percent and 14 percent, respectively) and nail-related AEs (69 percent and 57 percent, respectively; Grade 3/4: two percent and zero percent, respectively).3

"The updated findings from MonumenTAL-1 and data from the TRiMM-2 study are exciting, as they demonstrate the continued promise of T-cell redirecting therapies as single agents or in combination with standard-setting treatments in multiple myeloma," said Chris Heuck, M.D., Global Medical Head, Oncology, Janssen Research & Development, LLC. "At Janssen, we recognize that the future of multiple myeloma treatment lies in harnessing the power of combination therapies to target this complex disease, and the talquetamab results seen to date offer new hope to patients in need of additional treatment options."

About the MonumenTAL-1 Study
MonumenTAL-1 (Phase 1: NCT03399799, Phase 2: NCT04634552), is a Phase 1/2 single-arm, open-label, multicohort, multicenter dose-escalation study to evaluate the safety and efficacy of talquetamab in adults with relapsed or refractory multiple myeloma who received three or more prior lines of therapy, including a PI, an immunomodulatory agent, and an anti-CD38 monoclonal antibody.

Phase 1 of the study (NCT03399799) was conducted in 2 parts: dose escalation and dose expansion. It evaluated safety, tolerability, pharmacokinetics and preliminary antitumor activity of talquetamab administered to adult participants with relapsed or refractory multiple myeloma. Study criteria excluded patients who had an allogenic stem cell transplant within six months, Eastern Cooperative Oncology Group (ECOG) performance score of 2 or higher, known active central nervous system (CNS) involvement or clinical signs of meningeal involvement of multiple myeloma, or toxicities from previous anticancer therapies at Grade 2 or higher with the exception of alopecia or peripheral neuropathy.

Phase 2 of the study (NCT04634552) evaluated the efficacy of talquetamab in participants with relapsed or refractory multiple myeloma at the recommended Phase 2 doses, established at subcutaneous 0.8 mg/kg every two weeks and 0.4 mg/kg weekly, respectively, as measured by ORR.

The study also included 51 patients who were exposed to prior T-cell redirection therapy and had received at least three prior therapies. Prior T-cell redirection therapy was CAR-T cell therapy for 70.6 percent of patients and bispecific antibody treatment for 35.3 percent. With a median duration of follow-up of 14.8 months, ORR per IRC assessment was 64.7 percent.

About TRIMM-2 Study
The TRIMM-2 (NCT04108195) study is an ongoing Phase 2 study of DARZALEX FASPRO regimens in combination with talquetamab for the treatment of patients with multiple myeloma. The primary objectives of the TRiMM-2 study were to identify the Phase 2 dose (RP2D) for each component of the treatment combination (Part One); characterize the safety of the treatment combination at the RP2D (Part 2); and assess antitumor activity, pharmacokinetics and pharmacodynamics for the combination treatment (Part 3). Patients in the study (n=65) all had multiple myeloma and had received a minimum three prior lines of therapy or were double refractory to a proteasome inhibitor (PI) and an immunomodulatory agent; patients who had been exposed or refractory to an anti-CD38 therapy more than ninety days prior to the start of the trial were also included, as well as those refractory to anti-CD38 therapy.

About Talquetamab
Talquetamab is a bispecific T-cell engaging antibody that binds to the CD3 receptor expressed on the surface of T cells and G protein-coupled receptor class C group 5 member D (GPRC5D), a novel multiple myeloma target that does not shed and is highly expressed on the surface of multiple myeloma cells and non-malignant plasma cells, as well as some healthy tissues such as epithelial cells of the skin and tongue.6 CD3 is involved in activating T-cells, and GPRC5D is highly expressed on multiple myeloma cells.7,8

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

A Phase 1/2 clinical study of talquetamab for the treatment of relapsed or refractory multiple myeloma (NCT03399799) is currently underway. Talquetamab is also being explored in combination studies (NCT04586426, NCT04108195, NCT05050097, NCT05338775) and in a randomized Phase 3 study (NCT05455320). In January 2021, talquetamab was granted PRIME designation by the European Commission.

About DARZALEX FASPRO and DARZALEX
DARZALEX FASPRO (daratumumab and hyaluronidase-fihj) received U.S. FDA approval in May 2020 and is approved for eight indications in multiple myeloma (MM), three of which are for frontline treatment in newly diagnosed patients who are transplant eligible or ineligible.9 It is the only subcutaneous CD38-directed antibody approved to treat patients with MM. DARZALEX FASPRO is co-formulated with recombinant human hyaluronidase PH20 (rHuPH20), Halozyme’s ENHANZE drug delivery technology.

DARZALEX (daratumumab) received U.S. FDA approval in November 2015 and is approved in eight indications, three of which are in the frontline setting, including newly diagnosed patients who are transplant eligible and ineligible.10

DARZALEX is the first CD38-directed antibody approved to treat multiple myeloma.11 DARZALEX-based regimens have been used in the treatment of more than 360,000 patients worldwide and more than 68,000 patients in the U.S. alone.

In August 2012, Janssen Biotech, Inc. and Genmab A/S entered a worldwide agreement, which granted Janssen an exclusive license to develop, manufacture and commercialize daratumumab.

Since 2020, the National Comprehensive Cancer Network (NCCN) has recommended daratumumab based combination regimens for the treatment of newly diagnosed multiple myeloma and relapsed and refractory multiple myeloma.‡ For newly diagnosed multiple myeloma, the NCCN guidelines recommend daratumumab in combination with lenalidomide and dexamethasone as a Category 1 preferred regimen in non-transplant candidates; daratumumab in combination with bortezomib, melphalan, and prednisone as another recommended Category 1 regimen for non-transplant candidates; and daratumumab in combination with bortezomib, thalidomide and dexamethasone as a Category 2A regimen useful in certain circumstances for transplant candidates. In relapsed/refractory myeloma, four daratumumab regimens are listed as Category 1 preferred regimens for early relapses (1-3 prior therapies): daratumumab in combination with lenalidomide and dexamethasone; daratumumab in combination with bortezomib and dexamethasone; daratumumab in combination with carfilzomib and dexamethasone; and daratumumab in combination with pomalidomide and dexamethasone [after one prior therapy including lenalidomide and a proteasome inhibitor (PI)]. The NCCN also recommends daratumumab in combination with cyclophosphamide, bortezomib and dexamethasone as another Category 2A regimen for early relapses (1-3 prior therapies) and as monotherapy as a Category 2A regimen useful in certain circumstances for early relapse patients after at least three prior therapies, including a PI and an immunomodulatory agent, or for patients who are double refractory to a PI and an immunomodulatory agent.

For more information, visit www.DARZALEX.com.

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. In multiple myeloma, these plasma cells change, spread rapidly and replace normal cells in the bone marrow with tumors. Multiple myeloma is the third most common blood cancer and remains an incurable disease. In 2023, it is estimated that more than 35,000 people will be diagnosed with multiple myeloma in the U.S. and more than 12,000 people will die from the disease. People living with multiple myeloma have a 5-year relative survival rate of 53 percent. While some people diagnosed with multiple myeloma initially have no symptoms, most patients are diagnosed due to symptoms that can include bone fracture or pain, low red blood cell counts, tiredness, high calcium levels, kidney problems or infections.

DARZALEX IMPORTANT SAFETY INFORMATION 

INDICATIONS
DARZALEX (daratumumab) is indicated for the treatment of adult patients with multiple myeloma:

In combination with lenalidomide and dexamethasone in newly diagnosed patients who are ineligible for autologous stem cell transplant and in patients with relapsed or refractory multiple myeloma who have received at least one prior therapy
In combination with bortezomib, melphalan, and prednisone in newly diagnosed patients who are ineligible for autologous stem cell transplant
In combination with bortezomib, thalidomide, and dexamethasone in newly diagnosed patients who are eligible for autologous stem cell transplant
In combination with bortezomib and dexamethasone in patients who have received at least one prior therapy
In combination with carfilzomib and dexamethasone in patients with relapsed or refractory multiple myeloma who have received one to three prior lines of therapy
In combination with pomalidomide and dexamethasone in patients who have received at least two prior therapies including lenalidomide and a proteasome inhibitor
As monotherapy in patients who have received at least three prior lines of therapy including a proteasome inhibitor (PI) and an immunomodulatory agent or who are double-refractory to a PI and an immunomodulatory agent
CONTRAINDICATIONS 
DARZALEX is contraindicated in patients with a history of severe hypersensitivity (e.g., anaphylactic reactions) to daratumumab or any of the components of the formulation.

WARNINGS AND PRECAUTIONS 
Infusion-Related Reactions 
DARZALEX can cause severe and/or serious infusion-related reactions including anaphylactic reactions. These reactions can be life-threatening, and fatal outcomes have been reported. In clinical trials (monotherapy and combination: N=2066), infusion-related reactions occurred in 37% of patients with the Week 1 (16 mg/kg) infusion, 2% with the Week 2 infusion, and cumulatively 6% with subsequent infusions. Less than 1% of patients had a Grade 3/4 infusion-related reaction at Week 2 or subsequent infusions. The median time to onset was 1.5 hours (range: 0 to 73 hours). Nearly all reactions occurred during infusion or within 4 hours of completing DARZALEX. Severe reactions have occurred, including bronchospasm, hypoxia, dyspnea, hypertension, tachycardia, headache, laryngeal edema, pulmonary edema, and ocular adverse reactions, including choroidal effusion, acute myopia, and acute angle closure glaucoma. Signs and symptoms may include respiratory symptoms, such as nasal congestion, cough, throat irritation, as well as chills, vomiting, and nausea. Less common signs and symptoms were wheezing, allergic rhinitis, pyrexia, chest discomfort, pruritus, hypotension and blurred vision. 

When DARZALEX dosing was interrupted in the setting of ASCT (CASSIOPEIA) for a median of 3.75 months (range: 2.4 to 6.9 months), upon re-initiation of DARZALEX, the incidence of infusion-related reactions was 11% for the first infusion following ASCT. Infusion-related reactions occurring at re-initiation of DARZALEX following ASCT were consistent in terms of symptoms and severity (Grade 3 or 4: <1%) with those reported in previous studies at Week 2 or subsequent infusions. In EQUULEUS, patients receiving combination treatment (n=97) were administered the first 16 mg/kg dose at Week 1 split over two days, i.e., 8 mg/kg on Day 1 and Day 2, respectively. The incidence of any grade infusion-related reactions was 42%, with 36% of patients experiencing infusion-related reactions on Day 1 of Week 1, 4% on Day 2 of Week 1, and 8% with subsequent infusions.

Pre-medicate patients with antihistamines, antipyretics, and corticosteroids. Frequently monitor patients during the entire infusion. Interrupt DARZALEX infusion for reactions of any severity and institute medical management as needed. Permanently discontinue DARZALEX therapy if an anaphylactic reaction or life-threatening (Grade 4) reaction occurs and institute appropriate emergency care. For patients with Grade 1, 2, or 3 reactions, reduce the infusion rate when re-starting the infusion.  

To reduce the risk of delayed infusion-related reactions, administer oral corticosteroids to all patients following DARZALEX infusions. Patients with a history of chronic obstructive pulmonary disease may require additional post-infusion medications to manage respiratory complications. Consider prescribing short- and long-acting bronchodilators and inhaled corticosteroids for patients with chronic obstructive pulmonary disease. 

Ocular adverse reactions, including acute myopia and narrowing of the anterior chamber angle due to ciliochoroidal effusions with potential for increased intraocular pressure or glaucoma, have occurred with DARZALEX infusion. If ocular symptoms occur, interrupt DARZALEX infusion and seek immediate ophthalmologic evaluation prior to restarting DARZALEX. 

Interference With Serological Testing 
Daratumumab binds to CD38 on red blood cells (RBCs) and results in a positive indirect antiglobulin test (indirect Coombs test). Daratumumab-mediated positive indirect antiglobulin test may persist for up to 6 months after the last daratumumab infusion. Daratumumab bound to RBCs masks detection of antibodies to minor antigens in the patient’s serum. The determination of a patient’s ABO and Rh blood type is not impacted. Notify blood transfusion centers of this interference with serological testing and inform blood banks that a patient has received DARZALEX. Type and screen patients prior to starting DARZALEX. 

Neutropenia and Thrombocytopenia 
DARZALEX may increase neutropenia and thrombocytopenia induced by background therapy. Monitor complete blood cell counts periodically during treatment according to manufacturer’s prescribing information for background therapies. Monitor patients with neutropenia for signs of infection. Consider withholding DARZALEX until recovery of neutrophils or for recovery of platelets.

Interference With Determination of Complete Response 
Daratumumab is a human immunoglobulin G (IgG) kappa monoclonal antibody that can be detected on both the serum protein electrophoresis (SPE) and immunofixation (IFE) assays used for the clinical monitoring of endogenous M-protein. This interference can impact the determination of complete response and of disease progression in some patients with IgG kappa myeloma protein. 

Embryo-Fetal Toxicity 
Based on the mechanism of action, DARZALEX can cause fetal harm when administered to a pregnant woman. DARZALEX may cause depletion of fetal immune cells and decreased bone density. Advise pregnant women of the potential risk to a fetus. Advise females with reproductive potential to use effective contraception during treatment with DARZALEX and for 3 months after the last dose. 

The combination of DARZALEX with lenalidomide, pomalidomide, or thalidomide is contraindicated in pregnant women because lenalidomide, pomalidomide, and thalidomide may cause birth defects and death of the unborn child. Refer to the lenalidomide, pomalidomide, or thalidomide prescribing information on use during pregnancy. 

ADVERSE REACTIONS 
The most frequently reported adverse reactions (incidence ≥20%) were: upper respiratory infection, neutropenia, infusion-related reactions, thrombocytopenia, diarrhea, constipation, anemia, peripheral sensory neuropathy, fatigue, peripheral edema, nausea, cough, pyrexia, dyspnea, and asthenia. The most common hematologic laboratory abnormalities (≥40%) with DARZALEX are: neutropenia, lymphopenia, thrombocytopenia, leukopenia, and anemia.

Please click here to see the full Prescribing Information.

Sutro Biopharma Announces Oral Presentation at ASCO 2023 Featuring Data for Luveltamab Tazevibulin from the Phase 1 Dose-Expansion Study in Ovarian Cancer and the Initiation of the Phase 2/3 Pivotal Study REFRaME-O1

On June 3, 2023 Sutro Biopharma, Inc. (Sutro or the Company) (NASDAQ: STRO), a clinical-stage oncology company pioneering site-specific and novel-format antibody drug conjugates (ADCs), reported that results from a Phase 1 dose-expansion study of luveltamab tazevibulin (luvelta), a novel Folate receptor alpha (FolRα)-targeting ADC, in patients with advanced ovarian cancer were featured in an oral presentation at the 2023 American Society of Clinical Oncology (ASCO) (Free ASCO Whitepaper) (ASCO 2023) Annual Meeting in Chicago, IL (Press release, Sutro Biopharma, JUN 3, 2023, View Source [SID1234632429]). In parallel, the Company continues to advance the clinical development of luvelta and announced that sites are now open for enrollment in REFRaME-O1, the pivotal Phase 2/3 study for patients with platinum-resistant ovarian cancer.

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!

Data from the Phase 1 dose-expansion study of luvelta were presented by co-Principal Investigator, Ana Oaknin, M.D., Ph.D., Head of the Gynecological Tumor Unit and Attending Physician at the Vall d’Hebron University Hospital in Barcelona and Principal Clinical Investigator of the Gynecological Malignancies Group at the Vall d’Hebron Institute of Oncology. Consistent with data reported in January 2023, luvelta demonstrated substantial clinical benefit in FolRα-selected patients, defined by Tumor Proportion Score (TPS) of >25%, irrespective of staining intensity, which represents approximately 80% of the advanced ovarian cancer patient population.

When focusing on patients with a FolRα expression level above 25% (Tumor Proportion Score, or TPS >25% and regardless of staining intensity), the efficacy outcomes exhibited a 37.5% ORR, a median DOR of 5.5 months, and a median PFS of 6.1 months. Notably, at the higher starting dose level of 5.2 mg/kg, these patients experienced even higher response rates, with a 43.8% ORR, a median DOR of 5.4 months, and a median PFS of 6.6 months. Responses were seen in FolRα expressing patients with TPS >25%, addressing patients who may not be eligible for other approved therapies targeting FolRα.

"I am encouraged by the preliminary efficacy, durability, and favorable safety profile observed in this study, which signify the potential of luveltamab tazevibulin as a promising therapeutic option for patients with ovarian cancer who are not well supported by current standard of care," commented Dr. Oaknin. "The population of ovarian cancer patients that may benefit from treatment with luveltamab tazevibulin represents a significant unmet medical need globally."

"We are excited to share these promising data in an oral presentation at ASCO (Free ASCO Whitepaper), a globally respected oncology conference, which demonstrate the meaningful clinical benefit that luvelta may offer to ovarian cancer patients across a broad range of heterogeneity in FolRα-expression," said Anne Borgman, M.D., Sutro’s Chief Medical Officer. "On the heels of these positive results, we are thrilled that REFRaME, our pivotal Phase 2/3 trial, is officially underway. From the clinical and nonclinical data gathered, we maintain our positive outlook that luvelta could potentially serve multiple additional indications where patients express FolRα."

About Luveltamab Tazevibulin
Luveltamab tazevibulin, abbreviated as "luvelta" and formerly known as STRO-002, is a FolRα-targeting antibody-drug conjugate (ADC) designed to treat a broad range of patients with ovarian cancer, including those with lower FolRα-expression who are not eligible for approved treatment options targeting FolRα. Developed and manufactured with Sutro’s cell-free XpressCF platform, luvelta is a homogeneous ADC with four hemiasterlin cytotoxins per antibody, precisely positioned to efficiently deliver to the tumor while ensuring systemic stability after dosing. Sutro recently initiated REFRaME, a Phase 2/3 registration-directed study for patients with platinum-resistant ovarian cancer. The company has ongoing trials in patients with endometrial cancer and in combination with bevacizumab in patients with ovarian cancer. The company is also assessing the clinical path forward for CBF/GLIS2 acute myeloid leukemia, a rare subtype of pediatric cancer, as well as non-small cell lung cancer.

Gilead And Arcus Announce Anti-TIGIT Domvanalimab Continues To Demonstrate Consistent Improvement In Progression-Free Survival In Non-Small Cell Lung Cancer Study

On June 3, 2023 Gilead Sciences, Inc. (Nasdaq: GILD) and Arcus Biosciences, Inc. (NYSE: RCUS) reported updated results from an interim analysis of the ARC-7 study in patients with first-line, metastatic non-small cell lung cancer (NSCLC) with PD-L1 tumor proportion score (TPS) ≥50% without epidermal growth factor receptor (EGFR) or anaplastic lymphoma kinase (ALK) mutations. ARC-7 is the first Phase 2, randomized, open-label study evaluating the combinations of Fc-silent anti-TIGIT monoclonal antibody domvanalimab plus anti-PD-1 monoclonal antibody zimberelimab (doublet) and domvanalimab plus zimberelimab and etrumadenant, an A2a/b adenosine receptor antagonist (triplet), versus zimberelimab monotherapy (Press release, Gilead Sciences, JUN 3, 2023, View Source [SID1234632428]). These results will be presented today during the American Society of Clinical Oncology (ASCO) (Free ASCO Whitepaper) Plenary Series: Rapid Abstract Updates session by Melissa L. Johnson, M.D., Director, Lung Cancer Research, Sarah Cannon Research Institute, and Lead Investigator for the ARC-7 study.

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!

"Progression-free survival curves showed early separation of both domvanalimab-containing arms from the zimberelimab arm, which was consistently maintained, and supports the potential therapeutic benefit of inhibiting the TIGIT pathway," said Melissa L. Johnson, M.D., Director, Lung Cancer Research, Sarah Cannon Research Institute, and Lead Investigator for the ARC-7 study. "I was also encouraged by the consistency of meaningful improvements across other outcome measures for the domvanalimab-containing arms. I look forward to continuing to work with Gilead and Arcus on the dom-zim combinations."

At the time of data cutoff (DCO), February 7, 2023, safety and efficacy were evaluated in all patients randomized and treated (n=150). With a median follow-up time of approximately 18 months, both domvanalimab-containing study arms demonstrated sustained, clinically meaningful improvements in progression-free survival (PFS) compared to zimberelimab monotherapy, with a 33% reduction in risk of disease progression or death for the doublet and 28% for the triplet.

The efficacy data, including PFS and ORR, are summarized in the table below:


Endpoint

zimberelimab (Z)
monotherapy
(n=50)

domvanalimab +
zimberelimab
(DZ)
(n=50)

etrumadenant +
domvanalimab +
zimberelimab
(EDZ) (n=50)


Progression-Free Survival (PFS)


Median in Months (95% CI)

5.4 (2.7, 9.7)

9.3 (4.1, NE)

9.9 (4.8, 14.6)


Hazard Ratio* (95% CI)

0.67 (0.4, 1.13)

0.72 (0.63, 1.8)


Six-month PFS rate (95% CI)

45% (30, 59)

58% (43, 72)

62% (48, 76)


12-month PFS rate (95% CI)

25% (11, 40)

41% (26, 56)

44% (29, 59)


Objective Response Rate (ORR)


ORR+ Confirmed + Pending (95% CI)

15 (30%)

[17.9%, 44.6%]

20 (40%)++

[26.4%, 54.8%]

22 (44%)

[30%, 58.7%]


Complete Response

1 (2%)

1 (2%)

0 (0%)


Partial Response Confirmed

14 (28%)

18 (36%)

22 (44%)


Partial Response Pending

0 (0%)

1 (2%)

0 (0%)


Stable Disease

16 (32%)

18 (36%)

16 (32%)


Progressive Disease

12 (24%)

4 (8%)

7 (14%)


Not Evaluable (NE)

7 (14%)

8 (16%)

5 (10%)

CI=Confidence Interval
*

Comparing DZ and EDZ arms to Z monotherapy.

+

Per RECIST 1.1

++

Across all arms, one participant in the DZ arm had a response pending confirmation, which was confirmed after DCO date.

Preliminary duration of response (DoR) analyses favor domvanalimab-containing arms, with median DoR (range, ‘+’: censored) as follows: Z: 13.2mo (+1.4-+19.4), DZ: not reached (2.8-+26.6), EDZ: 23.7mo (2.6-23.7).

As of the DCO, approximately twice as many participants remain on study treatment in the domvanalimab-containing arms compared to zimberelimab monotherapy [Z: (n=9), DZ: (n=17), EDZ: (n=20)].

Consistent ORR and PFS improvements were shown for the domvanalimab-containing arms in a post-hoc analysis of centrally confirmed PD-L1-high patients.

"At this analysis, the domvanalimab-containing study arms continued to show improved efficacy across multiple measures and both the doublet and triplet arms were generally well tolerated," said Dimitry S.A. Nuyten, M.D., Ph.D., Chief Medical Officer of Arcus Biosciences. "These data reinforce our confidence in the domvanalimab program."

"The ARC-7 proof-of-concept study has critically advanced our understanding of the activity of domvanalimab, the first Fc-silent anti-TIGIT monoclonal antibody in pivotal trials," said Bill Grossman, M.D., Ph.D., Senior Vice President, Therapeutic Area Head, Gilead Oncology. "We look forward to quickly advancing our four ongoing Phase 3 registrational programs in NSCLC and upper GI cancers."

No unexpected safety signals were observed across the three study arms at the time of DCO. The domvanalimab-containing study arms appeared to be generally well tolerated and showed an overall safety profile consistent with the known safety profiles of each individual molecule to date. Incidence of infusion-related reactions was low across all treatment arms: 4%, 4% and 12% for zimberelimab monotherapy and the domvanalimab-doublet and -triplet arms, respectively. The addition of domvanalimab to zimberelimab did not increase the incidence of infusion-related reactions, consistent with the Fc-silent design of domvanalimab.

Domvanalimab, zimberelimab and etrumadenant are investigational molecules. Neither Gilead nor Arcus has received approval from any regulatory authority for any use globally, and their safety and efficacy for the treatment of lung cancer have not been established.

About the ARC-7 Study

The ARC-7 study is the first Phase 2, multicenter, three-arm, randomized, open-label study evaluating the safety and efficacy of anti-TIGIT antibody domvanalimab plus anti-PD-1 antibody zimberelimab (doublet) versus domvanalimab plus zimberelimab and etrumadenant (triplet), an A2a/b adenosine receptor antagonist, versus zimberelimab monotherapy in 150 patients with first-line metastatic non-small cell lung cancer (NSCLC) with PD-L1 TPS ≥50% and no EGFR or ALK mutations. Patients are randomized 1:1:1 across the three study arms, and patients who progress on zimberelimab monotherapy may cross over to receive the triplet. At the time of this interim analysis, 150 patients had a median follow-up of 18.5 months. The co-primary endpoints are objective response rate and progression-free survival per Response Evaluation Criteria in Solid Tumors (RECIST 1.1). Secondary endpoints include duration of response, disease control rate, overall survival and safety. ARC-7 is a proof-of-concept study to assess the safety and efficacy of domvanalimab-containing study arms over zimberelimab monotherapy. More information about ARC-7 is available at: View Source

About Domvanalimab

Domvanalimab is the first Fc-silent investigational monoclonal antibody in pivotal trials that is designed to block and bind to the T-cell immunoreceptor with Ig and ITIM domains (TIGIT), a protein receptor on immune cells that acts as a brake on the immune response. Cancer cells can exploit TIGIT to avoid detection by the immune system. By binding to TIGIT, domvanalimab is expected to free up immune activating pathways and activate immune cells to attack and kill cancer cells. Domvanalimab has demonstrated complete receptor coverage on all TIGIT-expressing peripheral leukocytes.

Domvanalimab is being evaluated in four registrational Phase 3 studies across lung and gastrointestinal cancers, including: (1) ARC-10, evaluating domvanalimab plus zimberelimab versus pembrolizumab in first-line locally advanced or metastatic PD-L1 ≥50% NSCLC; (2) PACIFIC-8, being operationalized by AstraZeneca, evaluating domvanalimab plus durvalumab in unresectable Stage 3 NSCLC; (3) STAR-121, evaluating domvanalimab plus zimberelimab and chemotherapy versus pembrolizumab plus chemotherapy in first-line PD-L1-unselected NSCLC; and (4) STAR-221, evaluating domvanalimab plus zimberelimab and chemotherapy versus nivolumab plus chemotherapy in first-line locally advanced, unresectable or metastatic gastric, esophageal and gastro-esophageal junction adenocarcinomas.

Precigen Announces Positive Phase 1 Data for Off-the-Shelf PRGN-2009 AdenoVerse™ Immunotherapy Alone and in Combination with an Investigational Checkpoint Inhibitor in Patients with Recurrent/Metastatic HPV-associated Cancers

On June 3, 2023 Precigen, Inc. (Nasdaq: PGEN), a biopharmaceutical company specializing in the development of innovative gene and cell therapies to improve the lives of patients, reported positive clinical data from the Phase 1 study of the off-the-shelf (OTS) PRGN-2009 AdenoVerse immunotherapy alone and in combination with an investigational anti-PDL1/TGF-Beta Trap checkpoint inhibitor (bintrafusp alfa) in patients with recurrent/metastatic (R/M) HPV-associated cancers (clinical trial identifier: NCT04432597) at the 2023 American Society of Clinical Oncology (ASCO) (Free ASCO Whitepaper) Annual Meeting (Abstract# 2628) (Press release, Precigen, JUN 3, 2023, View Source [SID1234632427]).

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!

"Recurrent/metastatic HPV-associated cancers such as cervical, anal, and oropharyngeal are incurable by current therapies and there remains significant unmet need for new safe and effective treatments. Checkpoint inhibitors alone have shown some promise, but have not produced durable responses and many patients relapse or become resistant," said Helen Sabzevari, PhD, President and CEO of Precigen. "In our Phase 1 study, combining PRGN-2009 with checkpoint inhibition demonstrated a favorable safety profile and resulted in a 30% ORR with prolonged duration of responses in patients with heavily pre-treated HPV-associated cancers, including those who were checkpoint blockade resistant. We are highly encouraged by these Phase 1 data and, as we announced on May 31, we are moving rapidly to initiate a Phase 2 study to combine PRGN-2009 with a checkpoint inhibitor to further investigate safety and efficacy in recurrent or metastatic cervical cancer. This new trial is the second Phase 2 for PRGN-2009 AdenoVerse, adding to the ongoing Phase 2 in oropharyngeal squamous cell carcinoma."

PRGN-2009 is an OTS investigational immunotherapy utilizing the AdenoVerse platform designed to activate the immune system to recognize and target HPV-positive (HPV+) solid tumors. PRGN-2009 is a novel, replication-incompetent gorilla adenovirus targeting HPV 16/18. PRGN-2009 can be administered repeatedly leading to enhancement of T-cells without increasing neutralizing antibodies. PRGN-2009 is under development through a Cooperative Research and Development Agreement, or CRADA, within the Center for Immuno-Oncology (CIO), Center for Cancer Research (CCR), National Cancer Institute (NCI), part of the National Institutes of Health (NIH).

The Phase 1 trial is an open label, single-center study evaluating safety and response of PRGN-2009 as a monotherapy (Arm A) and in combination with bintrafusp alfa (Arm B) in previously treated adult patients with R/M HPV-associated cancers. In the monotherapy arm, patients (N = 6) enrolled in two sequential PRGN-2009 dose level cohorts, Dose Level 1 (1×1011 particle units (PU)) and Dose Level 2 (5×1011 PU) delivered via subcutaneous injection. In the combination arm, PRGN-2009 was administered at the recommended phase 2 dose (RP2D) in combination with bintrafusp alfa.

The primary objective of the study was to evaluate safety and RP2D of PRGN-2009 and safety of PRGN-2009 in combination with a checkpoint inhibitor. Secondary objectives included Objective Response Rate (ORR) (per RECIST 1.1), and progression free survival (PFS).

Patient Characteristics
Seventeen adult patients were enrolled in the Phase 1 study (Table 1). Patients received up to 20 doses of PRGN-2009 for a duration of 1.8 to 17.9 months in the monotherapy arm and 0.5 to 23.0 months in the combination arm. The median age in both arms was 61. The median number of prior lines of therapies in the metastatic setting was 2.5 for the monotherapy arm and 2 for the combination arm. All patients in the monotherapy arm (N=6) and 10 of 11 patients in the combination arm received prior immune checkpoint blockade (ICB) therapy.

Table 1. Patient Demographics and Clinical Characteristics

Monotherapy Arm (N=6)

Combination Arm (N=11)

Age, years (median, range)

61 (43-70)

61 (54-80)

Female, n (%)

6 (100)

3 (27)

Tumor types (n,%)

Oropharyngeal

Cervical

Anal

Vaginal

3 (50.0)

2 (33.3)

1 (16.7)

7 (63.6)

3 (27.3)

1 (9.1)

HPV status (n, %)

HPV16

HPV18

Other

N/A

3 (50)

2 (33.3)

1 (16.7)

9 (81.8)

1 (9.1)

1 (9.1)

Previous lines of therapy in metastatic setting, median (range)

2.5 (2-3)

2 (1-4)

ICB exposure, n (%)

Primary resistance

Secondary resistance

6 (100)

4 (66.7)

2 (33.3)

10 (90.9)

5 (50)

5 (50)

Safety Data
PRGN-2009 treatment in both monotherapy and combination arms was safe and well-tolerated (Table 2). In both study arms, there was a low incidence of treatment-related adverse events (TRAEs) with only Grade 1 or 2 TRAEs in the monotherapy arm. The most common TRAEs in the monotherapy arm were injection site reactions, flu-like symptoms, fatigue and rash. In addition to these in the combination arm, patients also experienced Grade 1 or 2 epistaxis, headache, keratoacanthoma, fever, decreased lymphocyte count, anemia and oral hemorrhage. TRAEs reported in the combination arm were in line with the safety profile reported for bintrafusp alfa and only Grade 1 or 2 TRAEs were attributable to PRGN-2009 in the combination arm.

Table 2: Safety Data

Monotherapy Arm (N=6)

Combination Arm (N=11)

Treatment-related adverse events, n (%)

Grade 1-2

(all)

Grade 3-4

(all)

Grade 1-2

(≥10%)

Grade 3-4

(all)

Injection site reactions

4 (66.7)

0

9 (81.8)

0

Flu-like symptoms,

3 (50.0)

0

6 (54.5)

0

Fatigue

2 (33.3)

0

3 (27.3)

0

Rash, maculopapular

1 (16.7)

0

3 (27.3)

0

Epistaxis

0

0

3 (27.3) *

0

Headache

0

0

3 (27.3)

0

Keratoacanthoma

0

0

3 (27.3)*

0

Fever

0

0

2 (18.2)

0

Lymphocyte count, decreased

0

0

2 (18.2)*

0

Anemia

0

0

2 (18.2)*

0

Oral hemorrhage

0

0

2 (18.2)*

0

Duodenal Hemorrhage

0

0

0

2 (18.2)*†‡

Pharyngeal mucositis

0

0

0

1 (9.1) *

*Attributed to bintrafusp alfa; † both patients concurrently receiving NSAIDs; ‡ 1 patient died following refusal of standard supportive medical management measures (blood transfusion).

Clinical Activity
Tumor responses were observed in patients after treatment with PRGN-2009 in combination with bintrafusp alfa (Arm 1B), including in ICB-resistant patients (Table 3). PRGN-2009 combined with bintrafusp alfa resulted in a 30% ORR in patients with pretreated R/M HPV-associated cancers with prolonged duration of responses (Table 3, Figure 1). The majority of patients developed HPV-16 and/or HPV-18 specific immune responses after treatment with PRGN-2009 in both monotherapy and combination arms (Table 4) without the development of neutralizing antibodies (Figure 2).

Table 3: Best Response by Arm

Monotherapy Arm

Combination Arm

Evaluable patients, n

6

10

Best response, n

CR

1a

PR

2c,d

SD

4b

1

PD

2

6

ORR, % (95% CI)

30 (6.7-65.3)

a immune checkpoint blockade (ICB)-resistant; b 1 SD confirmed; c 1 PR confirmed; d 1 ICB-resistant, 1 TCR treatment-resistant; 2 patients treated beyond progression without delayed response; CI: confidence interval

Figure 1: Time to Response and Duration of Response to Treatment

Figure 1: Time to Response and Duration of Response to Treatment

Table 4: HPV-specific T-cell Immune Responses

Monotherapy Arm

Combination Arm

HPV-16, n/N (%)

5/6 (83)

7/10 (70)

HPV-18, n/N (%)

5/6 (83)

7/8 (88)

HPV-16 and/or HPV-18 n/N (%)

6/6 (100)

8/10 (80)

N: number of patients tested

Figure 2: Neutralizing Antibodies

Figure 2: Neutralizing Antibodies

Figure 2: Neutralizing Antibodies

Phase 2 Clinical Study in Newly Diagnosed Oropharyngeal Squamous Cell Carcinoma
The Phase 2 portion of the study is ongoing at the RP2D and enrollment was completed in the monotherapy arm with 20 evaluable patients with newly diagnosed oropharyngeal squamous cell carcinoma (OPSCC). An interim clinical data presentation from the Phase 2 monotherapy arm is expected in the second half of 2023.

Phase 2 Randomized Control Study in Recurrent or Metastatic Cervical Cancer
The Company recently announced that the US Food and Drug Administration (FDA) has cleared the Investigational New Drug (IND) application to initiate a Phase 2 study of the first-in-class PRGN-2009 OTS AdenoVerse immunotherapy in combination with pembrolizumab in patients with recurrent or metastatic cervical cancer. The Phase 2 randomized, open-label, two-arm, multicenter study will evaluate the efficacy and safety of PRGN-2009 in combination with pembrolizumab versus pembrolizumab monotherapy in patients with recurrent or metastatic cervical cancer who are pembrolizumab resistant.

AFFIMED PRESENTS AFM24 MONOTHERAPY DATA IN NON-SMALL CELL LUNG CANCER AT THE ANNUAL MEETING OF THE AMERICAN SOCIETY OF CLINICAL ONCOLOGY AND PROVIDES STRATEGIC UPDATE ON AFM24 DEVELOPMENT PLAN

On June 3, 2023 Affimed N.V. (Nasdaq: AFMD) ("Affimed", or the "Company"), a clinical-stage immuno-oncology company committed to giving patients back their innate ability to fight cancer, reported the presentation at the American Society of Clinical Oncology (ASCO) (Free ASCO Whitepaper) Annual Meeting of safety and efficacy data from the EGFR mutant NSCLC expansion cohort of its ongoing phase 1/2 study investigating innate cell engager (ICE) AFM24 as monotherapy (Press release, Affimed, JUN 3, 2023, View Source [SID1234632426]). The data included 15 evaluable patients and showed encouraging early signs of clinical activity including confirmed partial responses and durable stable disease. EGFR mutant NSCLC is a very aggressive and resistant tumor type in which most classical NSCLC treatments achieve limited to no clinical activity, especially in more advanced patients. Affimed’s innate cell engager AFM24 aims to reactivate the innate and consequently the adaptive immune system to recognize and destroy EGFR mutant 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!

"The treatment options for patients with advanced solid tumors and EGFR alterations are limited by the development of resistance to existing EGFR targeting therapies and by modest activity of checkpoint inhibitors. The results we have seen with AFM24 monotherapy in an expansion cohort of advanced, refractory NSCLC patients with EGFR mutations indicate that activating the innate immune pathway in this patient population results in anti-tumor activity and may offer a novel therapeutic approach. Given the underlying molecular pathways, we believe that AFM24 could potentially enable checkpoint inhibitors to achieve a positive effect," said Dr. Anthony El-Khoueiry, Associate Director of Clinical Research at USC Norris Comprehensive Cancer Center and principal investigator of the AFM24 studies.

The AFM24 EGFR mutant NSCLC cohort is part of the AFM24-101 open-label, non-randomized, multi-center, phase 1/2a study (NCT04259450) investigating the safety, tolerability, and preliminary efficacy of AFM24 monotherapy in patients with advanced or metastatic EGFR+ solid tumors. Other cohorts being investigated included colorectal cancer (CRC) and renal carcinoma (RCC).

At the planned interim analysis, 15 patients with EGFR mutant NSCLC and a median of 2 prior lines of therapy had been treated with a median of 11 doses of AFM24. As of the cut-off date, the data showed clinical activity and signals of anti-tumor activity in 7 out of 15 heavily pre-treated patients, including two confirmed partial responses and five patients with stable disease (SD) resulting in an objective response rate of 13% and a disease control rate of 47%. All patients with stable disease were progression free for at least 3.5 months, with one patient exhibiting ongoing SD for more than 8 months. A reduction in tumor burden was observed in five of 13 patients (38%) with available baseline and subsequent tumor assessments based on RECIST criteria. All patients showed a well-managed safety profile with the majority exhibiting mild-to-moderate treatment-related adverse events in-line with previous findings, highlighting the well-managed safety profile that makes AFM24 a candidate for combination approaches. One Grade 5 (pneumonitis) adverse event was reported in a patient with progressive disease and multiple comorbidities; however, since relation to AFM24 could not be ruled out it is deemed treatment related. Although the formal continuation criteria for the cohort were not met, these data provide proof of concept that targeting NK cells can induce remission in patients with especially hard-to-treat solid tumors.

"We believe that AFM24 can be an important addition to the treatment armamentarium for addressing EGFR mutant tumors as the early anti-tumor effects support further evaluation in a combination setting with the goal of achieving meaningful patient benefit. That is why we are adding an EGFR mutant NSCLC cohort to our ongoing phase 1/2 study in combination with Roche’s PD-L1 checkpoint inhibitor atezolizumab," said Dr. Andreas Harstrick, Chief Medical Officer at Affimed. "Our broad AFM24 program aimed at identifying the right therapeutic settings and indications, and we believe that the data generated to date allow us to build the right path forward to maximize patient benefit."

STRATEGIC DEVELOPMENT OF AFM24
Based on the totality of the data accumulated for AFM24 to date, Affimed will focus its near-term development efforts on advancing AFM24 in combination with checkpoint inhibitors as part of its ongoing AFM24-102 study to further investigate the synergies between AFM24 and atezolizumab. Enrollment in the AFM24-101 monotherapy study will be concluded. An expansion cohort investigating EGFR mutant NSCLC will be added to AFM24-102 based on the encouraging signals observed in AFM24-101. Enrollment for the AFM24-102 combination study is ongoing with early encouraging case studies from the dose escalation supporting the hypothesis of combining innate and adaptive immunotherapy approaches in EGFR-positive solid tumors. An initial data update from the dose escalation and expansion part of the study is expected in the second half of 2023.

AFM24 is also currently being investigated in combination with an autologous NK cell product together with NKGen Biotech. The dose escalation part of this study is ongoing and initial data are expected to be available in H2 2023. Affimed and NKGen have mutually decided to discontinue the study. In line with Affimed’s NK cell combination experience for AFM13, the Company will evaluate the best options to advance this project with an allogeneic off-the-shelf NK cell product which the Company expects to be better suited for combination with AFM24 in a highly advanced patient population.

The Company will provide further details, including data from all three AFM24 monotherapy cohorts, and guidance on the clinical development plan for AFM24 in a conference call and webcast scheduled today.

Conference Call and Webcast Information Affimed will host a conference call and webcast on June 3, 2023, at 6:00 p.m. CDT / 7:00 p.m. EDT to review the monotherapy data and provide a strategic update on the AFM24 program going forward.

The conference call will be available via phone and webcast. The live audio webcast of the call will be available in the "Webcasts" section on the "Investors" page of the Affimed website at View Source To access the call by phone, please use link: https://register.vevent.com/register/BIca5147f060da49d5963a0b00a7bc8a66 and you will be provided with dial-in details and a pin number.

Note: To avoid delays, we encourage participants to dial into the conference call 15 minutes ahead of the scheduled start time. A replay of the webcast will be accessible at the same link for 30 days following the call.

More details about the programs for the ASCO (Free ASCO Whitepaper) Annual Meetings are available online at www.asco.org

About AFM24
AFM24 is a tetravalent, bispecific innate cell engager (ICE) that activates the innate immune system by binding to CD16A on innate immune cells and EGFR, a protein widely expressed on solid tumors, to kill cancer cells. Generated by Affimed’s fit-for-purpose ROCK platform, AFM24 represents a distinctive mechanism of action that uses EGFR as a docking site to engage innate immune cells for tumor cell killing through antibody-dependent cellular cytotoxicity and antibody-dependent cellular phagocytosis.

Affimed is evaluating AFM24 as monotherapy and in combinations with other cancer treatments in patients with advanced EGFR-expressing solid malignancies whose disease has progressed after treatment with previous anticancer therapies.

AFM24-101, a monotherapy, first-in-human phase 1/2a open-label, is a non-randomized, multi-center, multiple ascending dose escalation and expansion study. Additional details may be found at www.clinicaltrials.gov using the identifier NCT04259450.

AFM24 is also being evaluated in a phase 1/2a study in combination with Roche’s PD-L1 checkpoint inhibitor atezolizumab (AFM24-102, NCT05109442).

Furthermore, Affimed and NKGen Biotech are investigating AFM24 in combination with NKGen Biotech’s NK cell SNK01 in a phase 1/2a study (AFM24-103, NCT05099549).