Rgenix Presents Data on Phase I Clinical Activity For RGX-104 at the 2018 ASCO Annual Meeting

On June 4, 2018 Rgenix, Inc., a clinical stage biopharmaceutical company developing first-in-class small molecule and antibody cancer therapeutics, reported it is presenting data on the safety, pharmacodynamics, and clinical activity from the dose escalation stage of the ongoing Phase Ia/b trial of RGX-104, an oral small molecule immunotherapy that targets the liver X receptor (LXR) (Press release, Rgenix, JUN 4, 2018, View Source [SID1234527164]).

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

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

                  Schedule Your 30 min Free Demo!

In a poster presentation of an abstract accepted for the 2018 Annual Meeting of the American Society of Clinical Oncology (ASCO) (Free ASCO Whitepaper), "Pharmacodynamic and clinical activity of RGX-104, a first-in-class immunotherapy targeting the liver-X nuclear hormone receptor (LXR), in patients with refractory malignancies", Rgenix showed the first-in-class compound to be capable of generating immunologic and anti-tumor activity.

RGX-104 is a small-molecule LXR agonist that modulates innate immunity by activating the ApoE gene. In murine models, the small molecule depletes myeloid derived suppressor cells (MDSCs) and stimulates dendritic cells (DCs), activating anti-tumor immunity as a single agent as well as in combination with adoptive T cell therapy or checkpoint inhibitors. The Phase 1a/b trial in progress is studying the therapy with regards to safety, efficacy, pharmacokinetics and pharmacodynamics. A total of 26 patients with a broad array of tumors have received RGX-104 at a range of dose levels and frequency as part of the dose escalation stage of the study.

RGX-104 was well tolerated across dose cohorts, with hyperlipidemia – an on target effect of LXR agonism – representing the most common adverse event. Robust ApoE target gene engagement was observed in patients, along with substantial MDSC depletion and DC stimulation in 12 of 17 evaluable patients. Activation of circulating PD-1+ T cells was observed in 11 of the 12 patients that experienced MDSC depletion.

One patient with a high-grade neuroendocrine malignancy with small cell features had a confirmed radiographic partial response with a 53% reduction in index hepatic metastases at the 160 mg BID dose. This response was associated with a greater than 12-fold increase in activated PD-1+ T cells. Additionally, seven patients had stable disease for durations of 8-16 weeks. The dose of 160 mg BID was chosen as the Recommended Phase 2 Dose, with robust pharmacodynamic effects on ApoE expression and relevant immune cell populations.

Masoud Tavazoie, MD, PhD, and Chief Executive Officer of Rgenix, said, "Today’s presentation illustrates the promise of our lead clinical candidate RGX-104. It enables us to move forward with our plans to study the compound in expansion cohorts as a single agent as well as in combination with a PD-1 inhibitor in patients with both checkpoint-inhibitor refractory and naïve tumors." Escalation and Expansion cohorts in the Phase 1b stage of the clinical trial are currently enrolling patients with epithelial ovarian carcinoma (EOC), melanoma, non-small cell lung cancer (NSCLC), small cell lung cancer (SCLC), renal cell cancer (RCC), bladder cancer (BLC), and triple negative breast cancer (TNBC).

Pierre Fabre and Its Partner Array BioPharma Announce Additional Median Overall Survival Results of Encorafenib and Binimetinib in Patients with Braf-Mutant Advanced Melanoma

On June 4, 2018 Pierre Fabre and its partner Array BioPharma Inc. reported updated results from the Phase 3 COLUMBUS trial in BRAF-mutant advanced melanoma (Press release, Array BioPharma, JUN 4, 2018, View Source [SID1234527163]). The results showed median overall survival (mOS) was 33.6 months for patients treated with the combination of encorafenib and binimetinib compared with 16.9 months for patients treated with vemurafenib as a monotherapy. The combination reduced the risk of death compared with vemurafenib monotherapy (hazard ratio [HR] of 0.61 [95% CI: 0.47–0.79], p<0.0001]. The observed efficacy of vemurafenib in the control arm is also consistent with historical data, providing an additional benchmark for validating the patient population and results observed in COLUMBUS.1,2 Further, the two-year OS with the combination therapy was 58%. These results will be part of an oral presentation today at the American Society of Clinical Oncology (ASCO) (Free ASCO Whitepaper) annual meeting in Chicago, Illinois, and have been selected for the "Best of ASCO (Free ASCO Whitepaper)" program.

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!

"These data indicate that regardless of treatment group, the use of subsequent immunotherapies was similar, and therefore indicate that post-trial treatments are unlikely to have contributed to the OS results we’ve seen"

Tweet this
Importantly, the presentation included data showing limited use of post-trial immunotherapy, which is consistent with other published pivotal trials of BRAF and MEK inhibitors in BRAF-mutant advanced melanoma.1,3

"These data indicate that regardless of treatment group, the use of subsequent immunotherapies was similar, and therefore indicate that post-trial treatments are unlikely to have contributed to the OS results we’ve seen," said Professor Reinhard Dummer, University of Zurich, lead author and Vice-Chairman of the Department of Dermatology in the University Hospital of Zürich, Switzerland. "We’re pleased to present this data at ASCO (Free ASCO Whitepaper) which builds upon previous analyses of the COLUMBUS data and further support our belief that encorafenib and binimetinib could be a promising new treatment option for patients with BRAF-mutant advanced melanoma."

Additionally, the updated median progression-free survival (mPFS) results for patients treated with the combination of encorafenib and binimetinib remained consistent with what was previously reported: 14.9 months versus 7.3 months for patients treated with vemurafenib (HR=0.51 [95% CI 0.39–0.67]; p<0.0001).

As previously reported, the combination of encorafenib and binimetinib was generally well-tolerated. Grade 3–4 adverse events (AEs) that occurred in more than 5% of patients receiving the combination were increased gamma-glutamyltransferase (GGT; 9%), increased blood creatine phosphokinase (CK; 7%) and hypertension (6%). The incidence of selected any grade AEs of special interest, defined based on toxicities commonly associated with commercially available BRAF+MEK inhibitor treatments, for patients receiving the combination of encorafenib and binimetinib included: rash (22%), serous retinopathy including retinal pigment epithelial detachment (20%), pyrexia (18%) and photosensitivity (5%). Full safety results of COLOMBUS Part 1 were published in The Lancet Oncology.

About Melanoma
Metastatic melanoma is the most serious and life-threatening type of skin cancer and is associated with low survival rates.4,5 There are about 200,000 new cases of melanoma diagnosed worldwide each year, approximately half of which have BRAF mutations, a key target in the treatment of metastatic melanoma.4,6,7,8

About COLUMBUS
The COLUMBUS trial (NCT01909453) is a two-part, international, randomized, open-label Phase 3 trial evaluating the efficacy and safety of the combination of encorafenib and binimetinib compared with vemurafenib and encorafenib monotherapy in 921 patients with locally advanced, unresectable or metastatic melanoma with BRAFV600 mutation. Prior immunotherapy treatment was allowed. Over 200 sites across North America, Europe, South America, Africa, Asia and Australia participated in the trial. Patients were randomized into two parts:

In Part 1, 577 patients were randomized 1:1:1 to receive encorafenib 450 mg daily + binimetinib 45 mg twice daily (COMBO450); encorafenib 300 mg daily (ENCO 300); or vemurafenib 960 mg twice daily as a monotherapy. The dose of encorafenib in the combination arm is 50% higher than the single-agent maximum tolerated dose of 300 mg. A higher dose of encorafenib was possible due to improved tolerability when combined with binimetinib. The primary endpoint for the COLUMBUS trial was a median progression-free survival (mPFS) comparison of the COMBO450 arm versus vemurafenib. mPFS is determined based on tumor assessment (RECIST version 1.1 criteria) by a Blinded Independent Central Review (BICR). Secondary endpoints include a comparison of the mPFS of COMBO450 arm to that of ENCO300 and a comparison of overall survival (OS) in patients treated in the COMBO450 arm to that of vemurafenib alone. Results from Part 1 of the COLUMBUS trial previously published in The Lancet Oncology earlier this year (online March 2018, print May 2018), showed that COMBO450 more than doubled mPFS in patients with advanced BRAF-mutant melanoma, with a mPFS of 14.9 months compared with 7.3 months observed with vemurafenib (HR 0.54 [95% CI 0.41-0.71], p<0.0001). In the secondary mPFS comparison of COMBO450 to ENCO300, ENCO300 demonstrated a mPFS of 9.6 months (HR 0.75 [95% CI 0.56-1.00], p=0.051).
In Part 2, 344 patients were randomized 3:1 to receive encorafenib 300 mg daily plus binimetinib 45 mg twice daily (COMBO300) or ENCO300. Part 2 was designed to provide additional data to help evaluate the contribution of binimetinib to the combination of encorafenib and binimetinib.
As the secondary endpoint comparison of mPFS between the COMBO450 arm and ENCO300 arm in Part 1 did not achieve statistical significance, the protocol-specified analysis of OS is descriptive.

About Encorafenib and Binimetinib
BRAF and MEK are key protein kinases in the MAPK signaling pathway (RAS-RAF-MEK-ERK). Research has shown this pathway regulates several key cellular activities, including proliferation, differentiation, survival and angiogenesis. Inappropriate activation of proteins in this pathway has been shown to occur in many cancers, including melanoma and colorectal cancer. Encorafenib is a late-stage small molecule BRAF inhibitor and binimetinib is a late-stage small molecule MEK inhibitor, both of which target key enzymes in this pathway. Encorafenib and binimetinib are being studied in clinical trials in advanced cancer patients, including the Phase 3 COLUMBUS trial and the Phase 3 BEACON CRC trial.

Pierre Fabre has exclusive rights to commercialize encorafenib and binimetinib in Europe, Asia, Latin America and Australia. Pierre Fabre’s development partner, Array BioPharma, has exclusive rights in the U.S. and Canada, and has granted Ono Pharmaceutical exclusive rights to commercialize both products in Japan and South Korea. Encorafenib and binimetinib are investigational medicines and are not currently approved in any country.

Kite Announces New Data Analyses for CAR T Therapy in Patients with Blood Cancers at the 2018 American Society of Clinical Oncology Meeting

On May 4, 2018 Kite, a Gilead Company (Nasdaq: GILD), reported new analyses from the ZUMA chimeric antigen receptor T (CAR T) cell therapy development program that are being presented at the 2018 American Society of Clinical Oncology (ASCO) (Free ASCO Whitepaper) Annual Meeting in Chicago (Press release, Kite Pharma, MAY 4, 2018, View Source [SID1234527162]). The results include analyses of the ZUMA-1 study of Yescarta (axicabtagene ciloleucel) in adult patients with refractory large B-cell lymphoma showing that response status may predict rates of progression-free survival (PFS) (Abstract #3003) and that treatment responses were consistent across prior lines of therapy (Abstract #3039). Additionally, an analysis of the ZUMA-3 study evaluating investigational KTE-C19 for the treatment of adult patients with relapsed or refractory acute lymphoblastic leukemia (ALL) showed that patients experienced manageable safety and encouraging efficacy irrespective of prior blinatumomab use (Abstract #7006).

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!

"With the U.S. approval of Yescarta last year, we aim to transform the treatment of patients with refractory large B-cell lymphoma," said Alessandro Riva, MD, Gilead’s Executive Vice President, Oncology Therapeutics & Head, Cell Therapy. "We are also committed to studying Yescarta and other CD19-directed CAR T therapies for people with other relapsed or refractory blood cancers. Based on the strength of the ZUMA-1 data, we are now evaluating the potential of Yescarta in the second-line treatment setting in a Phase 3 study, ZUMA-7, and we continue to evaluate KTE-C19 in Phase 1/2 studies in ALL and other hematologic cancers."

Yescarta was the first CAR T cell therapy to be approved by the U.S. Food and Drug Administration (FDA) for the treatment of adult patients with relapsed or refractory large B-cell lymphoma after two or more lines of systemic therapy, including diffuse large B-cell lymphoma (DLBCL) not otherwise specified, primary mediastinal large B-cell lymphoma, high grade B-cell lymphoma and DLBCL arising from follicular lymphoma. Yescarta is not indicated for the treatment of patients with primary central nervous system lymphoma.

Yescarta has a Boxed Warning in its product label and an associated Risk Evaluation and Mitigation Strategy (REMS) regarding the risks of CRS and neurologic toxicities. Please see below for Important Safety Information.

A Marketing Authorization Application (MAA) for axicabtagene ciloleucel is currently under review with the European Medicines Agency (EMA).

Ongoing Responses, Response by Prior Lines of Therapy in ZUMA-1 (Abstracts #3003 and #3039)

Long-term ZUMA-1 follow-up data have shown an overall response rate (ORR) of 83 percent (n=84/101), including 58 percent (n=59/101) of patients with a complete response at a median follow-up of 15.1 months. In this long-term follow-up, Grade 3 or higher cytokine release syndrome (CRS) and neurologic events were seen in 12 percent and 29 percent of patients, respectively.

A new analysis of ZUMA-1 suggests that response status three months after infusion of Yescarta may be predictive of longer-term disease control. Of the 42 patients with complete response and nine with partial response at three months, the 12-month PFS rates were 79 percent and 78 percent, respectively. This abstract has also been selected for inclusion in the 2018 Best of ASCO (Free ASCO Whitepaper) program.

"We are encouraged by the strong long-term complete response rates in ZUMA-1, which represents a patient population that previously had few if any remaining treatment options," said Frederick L. Locke, MD, ZUMA-1 Co-Lead Investigator and Vice Chair of the Department of Blood and Marrow Transplant and Cellular Immunotherapy at Moffitt Cancer Center in Tampa, Florida. "Importantly, this new study analysis indicates that response status at three months is potentially predictive of prolonged PFS."

An additional ZUMA-1 analysis evaluated outcomes based on prior therapy the patients had received. The results indicate long-term clinical benefit for patients with refractory large B cell lymphoma, irrespective of the number of prior lines of therapy.

Rates of Response with Prior Blinatumomab Treatment in ZUMA-3 (Abstract #7006)

Phase 1 data for KTE-C19, an investigational CD19 CAR T cell therapy, presented at the 2017 Annual Meeting of the American Society of Hematology (ASH) (Free ASH Whitepaper) demonstrated high rates of complete response in adult patients with relapsed or refractory acute lymphoblastic leukemia (ALL). A new analysis of data from the ZUMA-3 study shows patients responded to KTE-C19 regardless of prior treatment with blinatumomab, an FDA-approved treatment for relapsed or refractory ALL. After eight or more weeks of follow-up, 63 percent (n=5/8) of patients with prior blinatumomab treatment and 80 percent (n=8/10) of patients without prior blinatumomab treatment had achieved a complete response or complete response with incomplete hematological recovery. Overall, 94 percent (n=17/18) of patients had minimal residual disease (MRD)-negative remission. KTE-C19 was also manufactured successfully in both groups, with similar product characteristics in terms of CD4/CD8 ratio and other measures.

"As a CD19/CD3 bispecific T cell antibody, the possible impact of prior blinatumomab use on the efficacy of KTE-C19 – a CD19-directed CAR T therapy – was an important question for exploration," said Bijal Shah, MD, ZUMA-3 investigator and medical oncologist, Moffitt Cancer Center. "We observed that prior blinatumomab use did not affect the manufacturing of efficacious product, and that high response rates were seen regardless of previous treatment with blinatumomab."

Grade 3 or higher CRS was seen in 27 percent of patients with prior blinatumomab and in 17 percent of patients without prior blinatumomab. Grade 3 or higher neurologic events were seen in 36 percent of patients with prior blinatumomab and 67 percent of patients without prior blinatumomab. A greater number of subjects in the blinatumomab-naïve group received the higher 1 × 106 cells/kg dose.

KTE-C19 for ALL is investigational and has not been proven safe or efficacious.

U.S. Important Safety Information for Yescarta

BOXED WARNING: CYTOKINE RELEASE SYNDROME AND NEUROLOGIC TOXICITIES

Cytokine Release Syndrome (CRS), including fatal or life-threatening reactions, occurred in patients receiving Yescarta. Do not administer Yescarta to patients with active infection or inflammatory disorders. Treat severe or life-threatening CRS with tocilizumab or tocilizumab and corticosteroids.
Neurologic toxicities, including fatal or life-threatening reactions, occurred in patients receiving Yescarta, including concurrently with CRS or after CRS resolution. Monitor for neurologic toxicities after treatment with Yescarta. Provide supportive care and/or corticosteroids as needed.
Yescarta is available only through a restricted program under a Risk Evaluation and Mitigation Strategy (REMS) called the Yescarta REMS.
CYTOKINE RELEASE SYNDROME (CRS): CRS occurred in 94% of patients, including 13% with ≥ Grade 3. Among patients who died after receiving Yescarta, 4 had ongoing CRS at death. The median time to onset was 2 days (range: 1-12 days) and median duration was 7 days (range: 2-58 days). Key manifestations include fever (78%), hypotension (41%), tachycardia (28%), hypoxia (22%), and chills (20%). Serious events that may be associated with CRS include cardiac arrhythmias (including atrial fibrillation and ventricular tachycardia), cardiac arrest, cardiac failure, renal insufficiency, capillary leak syndrome, hypotension, hypoxia, and hemophagocytic lymphohistiocytosis/macrophage activation syndrome. Ensure that 2 doses of tocilizumab are available prior to infusion of Yescarta. Monitor patients at least daily for 7 days at the certified healthcare facility following infusion for signs and symptoms of CRS. Monitor patients for signs or symptoms of CRS for 4 weeks after infusion. Counsel patients to seek immediate medical attention should signs or symptoms of CRS occur at any time. At the first sign of CRS, institute treatment with supportive care, tocilizumab or tocilizumab and corticosteroids as indicated.

NEUROLOGIC TOXICITIES: Neurologic toxicities occurred in 87% of patients. Ninety-eight percent of all neurologic toxicities occurred within the first 8 weeks, with a median time to onset of 4 days (range: 1-43 days) and a median duration of 17 days. Grade 3 or higher occurred in 31% of patients. The most common neurologic toxicities included encephalopathy (57%), headache (44%), tremor (31%), dizziness (21%), aphasia (18%), delirium (17%), insomnia (9%) and anxiety (9%). Prolonged encephalopathy lasting up to 173 days was noted. Serious events including leukoencephalopathy and seizures occurred with Yescarta. Fatal and serious cases of cerebral edema have occurred in patients treated with Yescarta. Monitor patients at least daily for 7 days at the certified healthcare facility following infusion for signs and symptoms of neurologic toxicities. Monitor patients for signs or symptoms of neurologic toxicities for 4 weeks after infusion and treat promptly.

YESCARTA REMS: Because of the risk of CRS and neurologic toxicities, Yescarta is available only through a restricted program under a Risk Evaluation and Mitigation Strategy (REMS) called the Yescarta REMS. The required components of the Yescarta REMS are: Healthcare facilities that dispense and administer Yescarta must be enrolled and comply with the REMS requirements. Certified healthcare facilities must have on-site, immediate access to tocilizumab, and ensure that a minimum of 2 doses of tocilizumab are available for each patient for infusion within 2 hours after Yescarta infusion, if needed for treatment of CRS. Certified healthcare facilities must ensure that healthcare providers who prescribe, dispense or administer Yescarta are trained about the management of CRS and neurologic toxicities. Further information is available at www.YESCARTAREMS.com or 1-844-454-KITE (5483).

HYPERSENSITIVITY REACTIONS: Allergic reactions may occur. Serious hypersensitivity reactions including anaphylaxis may be due to dimethyl sulfoxide (DMSO) or residual gentamicin in Yescarta.

SERIOUS INFECTIONS: Severe or life-threatening infections occurred. Infections (all grades) occurred in 38% of patients, and in 23% with ≥ Grade 3. Grade 3 or higher infections with an unspecified pathogen occurred in 16% of patients, bacterial infections in 9%, and viral infections in 4%. Yescarta should not be administered to patients with clinically significant active systemic infections. Monitor patients for signs and symptoms of infection before and after Yescarta infusion and treat appropriately. Administer prophylactic anti-microbials according to local guidelines. Febrile neutropenia was observed in 36% of patients and may be concurrent with CRS. In the event of febrile neutropenia, evaluate for infection and manage with broad spectrum antibiotics, fluids and other supportive care as medically indicated. Hepatitis B virus (HBV) reactivation, in some cases resulting in fulminant hepatitis, hepatic failure and death, can occur in patients treated with drugs directed against B cells. Perform screening for HBV, HCV, and HIV in accordance with clinical guidelines before collection of cells for manufacturing.

PROLONGED CYTOPENIAS: Patients may exhibit cytopenias for several weeks following lymphodepleting chemotherapy and Yescarta infusion. Grade 3 or higher cytopenias not resolved by Day 30 following Yescarta infusion occurred in 28% of patients and included thrombocytopenia (18%), neutropenia (15%), and anemia (3%). Monitor blood counts after Yescarta infusion.

HYPOGAMMAGLOBULINEMIA: B-cell aplasia and hypogammaglobulinemia can occur. Hypogammaglobulinemia occurred in 15% of patients. Monitor immunoglobulin levels after treatment and manage using infection precautions, antibiotic prophylaxis and immunoglobulin replacement. The safety of immunization with live viral vaccines during or following Yescarta treatment has not been studied. Vaccination with live virus vaccines is not recommended for at least 6 weeks prior to the start of lymphodepleting chemotherapy, during Yescarta treatment, and until immune recovery following treatment.

SECONDARY MALIGNANCIES: Patients may develop secondary malignancies. Monitor life-long for secondary malignancies. In the event that a secondary malignancy occurs, contact Kite at 1-844-454-KITE (5483) to obtain instructions on patient samples to collect for testing.

EFFECTS ON ABILITY TO DRIVE AND USE MACHINES: Due to the potential for neurologic events, including altered mental status or seizures, patients are at risk for altered or decreased consciousness or coordination in the 8 weeks following Yescarta infusion. Advise patients to refrain from driving and engaging in hazardous occupations or activities, such as operating heavy or potentially dangerous machinery, during this initial period.

ADVERSE REACTIONS: The most common adverse reactions (incidence ≥ 20%) include CRS, fever, hypotension, encephalopathy, tachycardia, fatigue, headache, decreased appetite, chills, diarrhea, febrile neutropenia, infections-pathogen unspecified, nausea, hypoxia, tremor, cough, vomiting, dizziness, constipation, and cardiac arrhythmias.

Please see accompanying full Prescribing Information, including BOXED WARNING and Medication Guide.

Ayala Pharmaceuticals Presents Phase 1b Data at the 2018 American Society of Clinical Oncology (ASCO) Annual Meeting for AL101, a Pan-Notch Inhibitor, in Patients with Locally Advanced or Metastatic Solid Tumors

On June 4, 2018 Ayala Pharmaceuticals, a clinical stage precision oncology biopharmaceutical company dedicated to developing novel targeted therapies for genomically defined cancers in patient populations with high unmet medical needs, reported results from the Phase 1b study of AL101 (formerly BMS-906024), a gamma secretase inhibitor that potently inhibits signaling downstream of Notch receptors (1, 2, 3 and 4) (Press release, Ayala Pharmaceuticals, JUN 4, 2018, View Source [SID1234527161]). These data were accepted for a poster presentation titled, "A phase I study of AL101, a pan-Notch inhibitor, in patients with locally advanced or metastatic solid tumors," at the 2018 American Society of Clinical Oncology (ASCO) (Free ASCO Whitepaper) Annual Meeting in Chicago. The poster was selected for a discussion session that will take place on Monday, June 4, 2018, from 3:00 p.m.-4:15 p.m. in Hall A, Room S406, McCormick Place.

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 primary objective of the study was to assess the safety and tolerability of multiple IV doses of AL101, and to establish the recommended Phase 2 dose. Secondary objectives were to assess the pharmacokinetics (PK) and pharmacodynamics (PD) of AL101 and its equally active metabolite after the first IV dose and after repeated doses. All study objectives were met.

AL101 is a best-in-class gamma secretase inhibitor that has demonstrated potent and selective inhibition downstream of all four Notch receptors in preclinical models. Based on these encouraging findings, a Phase 1 study was designed in advanced solid tumors to evaluate safety and tolerability as well as PK and PD of the compound.

Ninety-four patients were enrolled in the study and treated with one of two alternative regimens: Arm A (QW, n=83) and Arm B (Q2W, n=11) using a 3+3 design, with expansion at the maximum tolerated dose (MTD). Tumor types included adenoid cystic carcinoma (ACC), triple negative breast cancer (TNBC), non-small cell lung cancer (NSCLC) and selected other tumors with reported Notch activation. The defining dose-limiting toxicity (DLT) period was four weeks (4 doses QW or 2 doses Q2W). PD biomarkers of Notch activity, including HES1 mRNA, were evaluated in serial whole blood.

A MTD of 4 mg QW was established in the escalation phase and used in the expansion phase. The safety profile was consistent with that on target effects of Notch inhibition. The majority of adverse effects were low grade and manageable with protocol guidelines. Grade 3/4 events reported in >15% (all doses, Arm A) included: diarrhea 17 (20%), hypophosphatemia 31(37%), nausea 1 (1%), vomiting 4 (5%), hypokalemia 6 (7%).

Seven DLTs were reported in Arm A: four in patients receiving 6 mg, (Grade 3 vomiting, Grade 3 diarrhea, Grade 3 diarrhea/colonic ulcerations, Grade 3 diarrhea/Grade 4 dehydration) and in three patients receiving 8.4 mg (Recurrent Grade 3 infusion reaction, Grade 3 vomiting, Grade 5 hepatic failure). There were no DLTs in three DLT-evaluable patients at 6 mg QW during escalation, and once 8.4 mg QW was deemed above the MTD, 11 additional patients were enrolled at 6 mg (10 were DLT evaluable). There were no DLTs in seven DLT-evaluable patients receiving 4 mg QW.

Weekly dosing of AL101 led to continuous Notch inhibition as measured by HES 1 transcription at doses 4 mg QW and above. Clinical activity was demonstrated across different solid tumor types at the MTD as defined by RECIST v1.1: one complete response was observed in a patient with a gastroesophageal junction adenocarcinoma with two missense and one splice-site mutation in Notch 1. One partial response was observed in a patient with a desmoid tumor, and one PR was observed in a patient with an ACC, with mutated Notch 1.

"Ayala is dedicated to precision oncology, bringing forward targeted therapies for cancer patients with high unmet needs," said Roni Mamluk, Ph.D., Chief Executive Officer at Ayala Pharmaceuticals. "As we continue our clinical development plans for Ayala’s Phase 2 study in the second half of this year, we are particularly encouraged by AL101’s clinical activity seen to date and look forward to initiating trials in our lead indication, ACC, in patients with activated Notch pathway, an indication with no approved treatment and patients in need for a therapy."

Tarveda Therapeutics Announces Results from Phase 1 Study of PEN-221 Presented at the 2018 American Society for Clinical Oncology (ASCO) Annual Meeting

On June 4, 2018 Tarveda Therapeutics, Inc., a clinical stage biopharmaceutical company discovering and developing Pentarins as a new class of potent and selective medicines to treat a wide range of cancers, reported Phase 1 results that were presented at the American Society for Clinical Oncology (ASCO) (Free ASCO Whitepaper) Annual Meeting from an ongoing Phase 1/2a study of PEN-221 in patients with somatostatin receptor 2 (SSTR2)-expressing neuroendocrine tumors (NET) or small cell lung cancer (SCLC) (Press release, Tarveda Therapeutics, JUN 4, 2018, View Source [SID1234527160]). The results presented had a data cutoff date of February 23, 2018. As of April 11, 2018, four patients remained on 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!

"PEN-221, our lead Pentarin miniature drug conjugate, is designed to rapidly penetrate deep into solid tumors where it is highly selective for the somatostatin receptor 2 and accumulates its potent DM1 payload," said Richard Wooster, Ph.D., President of Research and Development and Chief Scientific Officer at Tarveda. "Based on the safety results and encouraging signs of antitumor activity seen in our Phase 1 dose escalation and safety study for PEN-221, we have initiated the Phase 2a portion of our Phase 1/2a trial of PEN-221 to explore its potential in treating patients with gastrointestinal midgut and pancreatic neuroendocrine tumors as well as small cell lung cancer."

The Phase 1 portion of the study was designed as a dose escalating study to assess safety, tolerability, pharmacokinetics, and preliminary antitumor activity of PEN-221 in patients with SSTR2-expressing advanced neuroendocrine or small cell lung cancers. The results show that PEN-221 appears to be well-tolerated with evidence of antitumor activity seen in multiple patients. The maximum tolerated dose (MTD) and the recommended Phase 2 dose (RP2D) was established as 18mg and will be further evaluated in the Phase 2a portion of the study.

"There is a very real need for new treatment options for patients living with neuroendocrine tumors and small cell lung cancer," said Melissa Johnson, M.D., Associate Director, Lung Cancer Research Program at Sarah Cannon Research Institute. "PEN-221, an SSTR2 directed drug conjugate linked to a DM1 cytotoxic payload, is an exciting and novel approach to treating patients with SSTR2 expressing tumors identified using biomarker imaging agents OctreoScan or Gallium-68 DOTATATE PET. PEN-221 was well tolerated by patients in this clinical trial. In addition, the encouraging signals of antitumor activity and prolonged stable disease further supports continuation of the study of PEN-221 in the Phase 2a trial."

Phase 1 Trial Design

PEN-221 was administered as a one-hour, intravenous infusion once every three weeks to escalating cohorts of two to six patients with SSTR2 expressing advanced solid tumors including advanced gastroenteropancreatic, lung, thymus or other neuroendocrine tumors or small cell lung cancers or large cell neuroendocrine tumors of the lung. Safety was assessed by vital sign measurements, physical examinations, neurological examinations, ECOG performance status, documentation of adverse events, clinical laboratory tests, and electrocardiography. Disease response was assessed by duration of response and standard RECIST criteria.

Safety Data

A safety analysis of all 23 patients demonstrated that PEN-221 was well-tolerated with no dose limiting toxicities up to 18mg. Two of three patients administered 25mg of PEN-221 experienced dose limiting toxicities that rapidly and fully resolved following treatment discontinuation.

The majority of treatment-related/treatment-emergent adverse events were mild (Grade 1) to moderate (Grade 2) with the most common being fatigue (48%), nausea (48%), diarrhea (44%), and peripheral neuropathy (26%). There were single reports of Grade 3 peripheral neuropathy, alanine aminotransferase (ALT) and aspartate aminotransferase (AST) increase and constipation at the 25mg dose. One patient at the 25mg dose experienced a Grade 3 drug induced liver injury.

The Maximum Tolerated Dose and Recommended Phase 2 Dose of PEN-221 is 18mg administered once every 3 weeks.

Efficacy data

There was preliminary evidence of antitumor activity:

Among 15 NET patients who were evaluable for response, 11 had stable disease (SD) at 9 weeks, of whom 8 were sustained for 18-45 weeks, including 2 ongoing patients with SD for 44 and 45 weeks at the time of this data review.
Target lesion shrinkage leading to minor responses at the time of this data review were observed in 3/7 patients who had either a GI or pancreatic NET (dose range 8-18 mg).
The only SCLC patient had SD for 12 weeks.
As of April 11, 2018, four patients remained in the Phase 1 portion of the study with stable disease of 5, 7, 12, 14 months respectively.

About PEN-221

PEN-221 is a miniature drug conjugate consisting of a peptide ligand, that is highly selective in targeting SSTR2, joined through a cleavable linker to the potent cytotoxic payload DM1. SSTR2 is overexpressed on the cell surface of a range of solid tumors including neuroendocrine tumors and small cell lung cancers. In non-clinical experiments, PEN-221 binds with high affinity and selectivity to SSTR2. On binding, PEN-221 triggers SSTR2 internalization resulting in the accumulation of the DM1 payload in tumor cells followed by cell cycle arrest and apoptosis.

PEN-221 is being evaluated in Phase 2a expansion cohorts enrolling patients with midgut neuroendocrine tumors, pancreatic neuroendocrine tumors, and small cell lung cancer (ClinicalTrials.gov Identifier: NCT02936323).

About Pentarins
Tarveda is developing Pentarins, potent and selective miniature drug conjugates with high affinity for specific cell surface and intracellular targets. Pentarins are engineered to bind to their tumor cell targets and provide sustained release of their potent therapeutic payloads deep into solid tumor tissue. Comprised of a targeting ligand conjugated to a potent cancer cell killing agent through a tuned chemical linker, Pentarins are designed to overcome the deficits of both larger antibody drug conjugates and small molecules that limit their therapeutic effectiveness against solid tumors. Together, the components of Tarveda’s Pentarins have distinct, yet synergistic, anticancer attributes: the small size of Pentarins allows for rapid and deep penetration into the tumor tissue, the ligand’s targeting ability allows for specific binding and retention in tumor cells, and the chemical linker is tuned to optimize the release of the potent, cell killing payload inside the cancer cells for efficacy.