Verseau Launches with $50 Million to Develop First-in-Class Macrophage-Targeted Immunotherapies and Appoints George Golumbeski as Chairman of the Board

On October 21, 2019 Verseau Therapeutics, Inc. ("Verseau") reported that launched with $50 million in financing from 20/20 HealthCare Partners, 3SBio, Alexandria Venture Investments, Highlight Capital, InHarv Partners Ltd., The Mark Foundation for Cancer Research and Yonghua Capital (Press release, Verseau Therapeutics, OCT 21, 2019, View Source [SID1234542369]). In addition, George Golumbeski, Ph.D., a champion of innovation and former Executive Vice President of Celgene, has been appointed Chairman of the Verseau Board of Directors. Verseau is developing novel, first-in-class immunotherapies that target modulation of macrophages, the master orchestrators of the immune system. The proceeds from the financing will support advancement of Verseau’s macrophage checkpoint modulator (MCM) programs to the clinic.

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!

Macrophages can adopt different functional roles in response to signals from their environment, including the ability to direct pro-inflammatory and anti-inflammatory immune responses. Verseau has licensed an siRNA delivery technology, a lipid nanoparticle, from the MIT laboratories of Verseau’s co-founders Dan Anderson, Ph.D. and Bob Langer, Ph.D. Verseau is using such delivery technologies as part of its all human translational system to discover and validate novel macrophage targets, creating an expansive pipeline of macrophage checkpoint modulators.

The lead program targeting PSGL-1 reprograms macrophages to a pro-inflammatory state, activates T-cells and attracts other immune cells to generate a coordinated and powerful antitumor response.

"Current immunotherapies can only provide clinical benefit in the ~25% of cancers that involve T-cell infiltration. By targeting macrophages, present in 75% of human tumors, we believe we can offer potential clinical benefits of immunotherapy to a large, underserved patient population. Macrophage modulation as monotherapy and in combination with other therapies could provide enhanced clinical benefit for patients," said Dr. Christine Bunt, Chief Executive Officer of Verseau.

"Using our proprietary discovery and validation platform, we identified PSGL-1, an adhesion molecule that is highly expressed on tumor-associated macrophages across most tumor types, as the target of our first-in-class MCM program," said Dr. Tatiana Novobrantseva, Co-Founder and Chief Scientific Officer of Verseau. "Our PSGL-1 MCM antibody is designed to reprogram inhibitory tumor-associated macrophages into anti-cancer immune response stimulators. Verseau has validated more than two dozen targets amenable to different therapeutic modalities, including monoclonal antibodies."

"The focus on myeloid cells as an avenue to broaden the therapeutic potential of immunotherapy is emerging quickly, and Verseau is positioned to make a significant impact on this field. The company has a strong understanding of myeloid biology, has done some elegant screening for novel myeloid targets, and now is advancing a broad portfolio of antibody drug candidates," said Dr. George Golumbeski, Chairman of the Board of Verseau. "The early data are impressive and suggest that macrophage-targeted therapeutics may become a significant advance in immunotherapy. I look forward to working with the Verseau team to build the company and to advance the pipeline of drug candidates."

Verseau has a strategic collaboration with 3SBio, a fully-integrated biotechnology company in China with market-leading biopharmaceutical franchises. Under the agreement, 3SBio will receive an exclusive license to develop and commercialize a select number of MCM antibodies for all human oncology indications in Greater China, including mainland China, Taiwan, Hong Kong and Macau ("Territory"). Verseau retains all global rights.

About PSGL-1
PSGL-1 (P-selectin glycoprotein ligand-1) is an adhesion molecule that is involved in immune cell trafficking in response to tissue injury or inflammation. Verseau discovered that modulation of PSGL-1 can lead to macrophage reprogramming. Proprietary PSGL-1 monoclonal antibodies induce tumor microenvironment activation, T-cell activation and naïve immune cell recruitment amounting to a coordinated immune attack on tumors. In patient-derived primary tumors, PSGL-1 antibodies demonstrate a greater inflammatory response compared to current immunotherapies in both PD-1 responsive and non-responsive tumors. Given the prominent role of PSGL-1 in many tumor types, Verseau has selected PSGL-1 as the lead macrophage checkpoint modulator (MCM) program for clinical development.

Innovent Biologics Announces Positive Updated Result from the Incyte-sponsored Phase II Trial of Pemigatinib in Patients with Previously Treated, Advanced Cholangiocarcinoma

On October 20, 2019 Innovent Biologics, Inc. ("Innovent" or "the Company") (HKEX: 01801), a world-class biopharmaceutical company that develops and commercializes high quality medicines for the treatment of oncology, autoimmune, metabolic and other major diseases, reported positive updated results from the Incyte-sponsored Phase 2 trial of pemigatinib in patients with previously treated, advanced cholangiocarcinoma which were presented at the European Society for Medical Oncology (ESMO) (Free ESMO Whitepaper) 2019 (Press release, Innovent Biologics, OCT 20, 2019, View Source [SID1234542371]).

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 December 2018, Innovent and Incyte entered into a strategic collaboration for three clinical-stage product candidates discovered and developed by Incyte, including pemigatinib (FGFR1/2/3 inhibitor), itacitinib (JAK1 inhibitor) and parsaclisib (PI3Kδ inhibitor). Under the terms of the agreement, Innovent has received the rights to develop and commercialize the three assets in hematology and oncology in Mainland China, Hong Kong, Macau and Taiwan.

The updated results from the Phase 2 FIGHT-202 trial evaluating pemigatinib, a selective fibroblast growth factor receptor (FGFR) inhibitor, as a treatment for patients with previously treated, locally advanced or metastatic cholangiocarcinoma presented at ESMO (Free ESMO Whitepaper) 2019 include the final result for the primary endpoint. In patients harboring FGFR2 fusions or rearrangements (Cohort A), pemigatinib monotherapy resulted in an overall response rate (ORR) of 36 percent (primary endpoint), and median progression free survival (PFS) of 6.9 months (secondary endpoint) with a median follow-up of 15 months. Pemigatinib was generally well tolerated.

The data support the planned submission of a New Drug Application (NDA) by Incyte to the U.S. Food and Drug Administration (FDA) for pemigatinib before the end of 2019.

Cholangiocarcinoma is a rare cancer that forms in the bile duct. It is classified based on its origin: intrahepatic cholangiocarcinoma (iCCA) occurs in the bile duct inside the liver and extrahepatic cholangiocarcinoma occurs in the bile duct outside the liver. Patients with cholangiocarcinoma are often diagnosed at a late or advanced stage when the prognosis is poor. The incidence of cholangiocarcinoma varies regionally and ranges between 0.3– 3.4 per 100,000 in North America and Europe. FGFR2 fusions or rearrangements occur almost exclusively in iCCA, where they are observed in 10-16 percent of patients.

Key Findings from FIGHT-202

Updated data presented at ESMO (Free ESMO Whitepaper) show that in patients with previously treated, locally advanced or metastatic cholangiocarcinoma with FGFR2 fusions or rearrangements (Cohort A, n=107), pemigatinib monotherapy resulted in a confirmed overall response rate (ORR) of 36 percent based on an independent central radiographic review, including 3 patients with a complete response (CR) and 35 patients with a partial response (PR). In these patients, the disease control rate (DCR) was 82 percent, median duration of response (DOR) was 7.5 months, and median progression free survival (PFS) was 6.9 months. Preliminary overall survival (OS) data were encouraging (median: 21.1 months) and follow-up will continue as these data are not yet mature.

FIGHT-202 Overall Response Rates (ORR). Durability of Response (DOR). Disease Control Rates (DCR) and Progression-Free Survival (PFS) by Patient Cohort

Cohort A

FGFR2 Fusions or Rearrangements

Cohort B

Other FGF/FGFR Genetic Alterations

Cohort C

No FGF/FGFR Genetic Alterations

(N=107)

(N=20)

(N=18)

ORR, % (95% CI)

36 (27-45)

0

0

3 CR (3)

0

0

Best OR, n(%)

35 PR (33)

0

0

50 SD (47)

8 SD (40)

4 SD (22)

Median DOR,

Months (95% CI)

7.5 (5.7-14.5)

DCR, % (95% CI)

82 (74-89)

40 (19-64)

22 (6-48)

Median PFS,

Months (95% CI)

6.9 (6.2-9.6)

2.1 (1.2-4.9)

1.7 (1.3-1.8)

Median OS,

Months(95% CI)

21.1 (14.8-NE)

6.7 (2.1-10.6)

4.0 (2.3-6.5)

NE: not evaluable

Note: One patient did not have confirmed FGF/FGFR status by central laboratory and was included in the safety analysis but was not assigned to any cohort for efficacy.

The safety analysis, including 146 patients, showed that pemigatinib was generally well tolerated. Grade 1 or 2 hyperphosphatemia, the most common treatment-emergent adverse event (TEAE; 60 percent), was managed with a low phosphate diet, phosphate binders and diuretics, or dose reduction or interruption. The most common Grade ≥3 TEAE was hypophosphatemia (12 percent); none of the cases was considered clinically significant or serious and none led to dose reduction or discontinuation. Serous retinal detachment was observed in 4 percent of patients (Grade ≥3, 1 percent) with none of the cases resulting in clinical sequelae.

About FIGHT-202

The FIGHT-202 Phase 2, open-label, multicenter study (NCT02924376) is evaluating the safety and efficacy of pemigatinib – a selective fibroblast growth factor receptor (FGFR) inhibitor – in adult (age ≥ 18 years) patients with previously treated, locally advanced or metastatic cholangiocarcinoma with documented FGF/FGFR status.

Patients were enrolled into one of three cohorts – Cohort A (FGFR2 fusions or rearrangements), Cohort B (other FGF/FGFR genetic alterations) or Cohort C (no FGF/FGFR genetic alterations). All patients received 13.5 mg pemigatinib orally once daily (QD) on a 21-day cycle (two weeks on/one week off) until radiological disease progression or unacceptable toxicity.

The primary endpoint of FIGHT-202 is overall response rate (ORR) in Cohort A, assessed by independent review per RECIST v1.1. Secondary endpoints include ORR in Cohorts B, A plus B, and C; progression free survival (PFS), overall survival (OS), duration of response (DOR), disease control rate (DCR) and safety in all cohorts.

For more information about FIGHT-202, visit:

View Source

About FIGHT

The FIGHT (FIbroblast Growth factor receptor in oncology and Hematology Trials) clinical trial program includes ongoing Phase 2 and 3 studies investigating safety and efficacy of pemigatinib therapy across several FGFR-driven malignancies. Phase 2 monotherapy studies include FIGHT-202, as well as FIGHT-201 investigating pemigatinib in patients with metastatic or surgically unresectable bladder cancer, including with activating FGFR3 mutations or fusions/rearrangements; FIGHT-203 in patients with myeloproliferative neoplasms with activating FGFR1 fusions/rearrangements; FIGHT-207 in patients with previously treated, locally-advanced/metastatic or surgically unresectable solid tumor malignancies harboring activating FGFR mutations or fusions/rearrangements, irrespective of tumor type. FIGHT-205 is a Phase 2 study investigating pemigatinib plus pembrolizumab combination therapy and pemigatinib monotherapy in patients with previously untreated, metastatic or unresectable bladder cancer harboring FGFR3 mutations or fusions/rearrangements who are not eligible to receive cisplatin. FIGHT-302 is a recently initiated Phase 3 study investigating pemigatinib as a first-line treatment for patients with cholangiocarcinoma with FGFR2 fusions or rearrangements.

About FGFR and Pemigatinib

Fibroblast growth factor receptors (FGFRs) play an important role in tumor cell proliferation and survival, migration and angiogenesis (the formation of new blood vessels). Activating fusions, rearrangements, translocations and gene amplifications in FGFRs are closely correlated with the development of various cancers.

Pemigatinib is a potent, selective, oral inhibitor of FGFR isoforms 1, 2 and 3 which, in preclinical studies, has demonstrated selective pharmacologic activity against cancer cells with FGFR alterations discovered and developed by Incyte. The U.S. Food and Drug Administration (FDA) has granted pemigatinib Breakthrough Therapy designation for the treatment of previously treated, advanced/metastatic or unresectable FGFR2 translocated cholangiocarcinoma. The FDA’s Breakthrough Therapy designation is designed to expedite the development and review of drugs for serious conditions that have shown encouraging early clinical results and may demonstrate substantial improvements over available medicines.

CHMP Grants Positive Opinion for Expanded Use of Janssen’s Darzalex®▼(daratumumab) for Patients with Newly Diagnosed Multiple Myeloma Who Are Transplant Ineligible

On October 18, 2019 The Janssen Pharmaceutical Companies of Johnson & Johnson reported that the Committee for Medicinal Products for Human Use (CHMP) of the European Medicines Agency (EMA) has recommended broadening the existing marketing authorisation for Darzalex (daratumumab) to include the use of daratumumab in combination with lenalidomide and dexamethasone (DRd) for patients with newly diagnosed multiple myeloma (NDMM) who are ineligible for autologous stem cell transplant (ASCT) (Press release, Janssen Pharmaceuticals, OCT 18, 2019, View Source [SID1234542368]).

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!

"As multiple myeloma can become more complex with each relapse, it is important that patients receive the latest treatment options with the goal of extending their first remission period," said Professor Thierry Facon, M.D., Service des Maladies du Sang, Hôpital Claude Huriez, Lille, France, and principal investigator of the MAIA study. "For newly diagnosed patients who are transplant ineligible, this regimen could be an important frontline therapy option and reinforces the consistent clinical profile of daratumumab."

This Positive Opinion is based on results from the Phase 3 MAIA (MMY3008) study, published in The New England Journal of Medicine,1 and presented at the 2018 American Society of Hematology (ASH) (Free ASH Whitepaper) Annual Meeting.

Additional information about the MAIA study can be found at www.ClinicalTrials.gov (NCT02252172).

"This recommendation marks an important step towards realising our ambition to improve outcomes for patients with multiple myeloma, right from diagnosis, especially for the majority of patients who are not eligible for transplant," said Craig Tendler, M.D., Vice President, Clinical Development and Global Medical Affairs, Oncology, Janssen Research & Development, LLC.

"Daratumumab has been used to treat more than 100,000 patients worldwide and we look forward to working with regulatory authorities to bring this important therapy to even more patients with multiple myeloma," adds Dr Patrick Laroche, Haematology Therapy Area Lead, Europe, Middle East and Africa (EMEA), Janssen-Cilag France.

This Opinion will now be reviewed by the European Commission (EC), which has the authority to grant final approval of the indications.

Professor Thierry Facon was the principal investigator in the MAIA study. He was not compensated for any media work.

#ENDS#

About the MAIA (NCT02252172) Trial2
The randomised, open-label, multicentre Phase 3 study included 737 NDMM patients ineligible for high-dose chemotherapy and ASCT aged 45-90 years old (median age of 73 years). Patients were randomised to receive either daratumumab-Rd or Rd alone in 28-day Cycles. In the daratumumab-Rd treatment arm, patients received daratumumab 16 (mg/kg) IV weekly for Cycles 1 – 2, every two weeks for Cycles 3 – 6 and every 4 weeks for Cycle 7 and thereafter. The primary endpoint was Progression-Free Survival (PFS), defined as the time from date of randomisation to either progressive disease (PD), or death, whichever occurred first. Patients in the daratumumab-Rd and Rd treatment arm received 25 mg of lenalidomide on Days 1 – 21 of each 28-day Cycle, and dexamethasone at 40 mg once a week for each Cycle. Patients in both treatment arms continued until disease progression or unacceptable toxicity.

About daratumumab
Daratumumab is a first-in-class3 biologic targeting CD38, a surface protein that is highly expressed across multiple myeloma cells, regardless of disease stage.4 Daratumumab is believed to induce tumour cell death through multiple immune-mediated mechanisms of action, including complement-dependent cytotoxicity (CDC), antibody-dependent cell-mediated cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP), as well as through apoptosis, in which a series of molecular steps in a cell lead to its death.5 A subset of myeloid derived suppressor cells (CD38+ MDSCs), CD38+ regulatory T cells (Tregs) and CD38+ B cells (Bregs) were decreased by daratumumab.5 Since launch, it is estimated that 100,000 patients have been treated with daratumumab worldwide.6 Daratumumab is being evaluated in a comprehensive clinical development programme across a range of treatment settings in multiple myeloma, such as in frontline and relapsed settings.7,8,9,10,11,12,13,14 Additional studies are ongoing or planned to assess its potential in other malignant and pre-malignant haematologic diseases in which CD38 is expressed, such as smouldering myeloma.15,16 For more information, please see www.clinicaltrials.gov.

The most frequent adverse reactions seen with daratumumab include infusion reactions, fatigue, nausea, diarrhoea, muscle spasms, pyrexia, cough, neutropenia, thrombocytopenia, anaemia, peripheral sensory neuropathy and upper respiratory tract infection.5

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

In August 2012, Janssen Biotech, Inc. and Genmab A/S entered a worldwide agreement, which granted Janssen an exclusive licence to develop, manufacture and commercialise daratumumab.17

About Multiple Myeloma
Multiple myeloma (MM) is an incurable blood cancer that starts in the bone marrow and is characterised by an excessive proliferation of plasma cells.18 In Europe, more than 48,200 people were diagnosed with MM in 2018, and more than 30,800 patients died.19 Almost 60 percent of patients with MM do not survive more than five years after diagnosis.20

Although treatment may result in remission, unfortunately, patients will most likely relapse as there is currently no cure.21 Refractory MM is when a patient’s disease progresses within 60 days of their last therapy.22,23 Relapsed cancer is when the disease has returned after a period of initial, partial or complete remission.24 While some patients with MM have no symptoms at all, most patients are diagnosed due to symptoms that can include bone problems, low blood counts, calcium elevation, kidney problems or infections.25 Patients who relapse after treatment with standard therapies, including proteasome inhibitors and immunomodulatory agents, have poor prognoses and few treatment options available.26

Daiichi Sankyo Receives Negative CHMP Opinion for FLT3 Inhibitor Quizartinib for Treatment of Patients with Relapsed/ Refractory FLT3-ITD AML

On October 18, 2019 Daiichi Sankyo Company, Limited (hereafter, Daiichi Sankyo) reported the Committee for Medicinal Products for Human Use (CHMP) of the European Medicines Agency (EMA) has adopted a negative opinion on the Marketing Authorization Application (MAA) for quizartinib for the treatment of adults with relapsed/refractory FLT3-ITD acute myeloid leukemia (AML) (Press release, Daiichi Sankyo, OCT 18, 2019, View Source [SID1234542366]).

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 CHMP opinion is based on the MAA submission of data from the global pivotal QuANTUM-R study of quizartinib. Results from QuANTUM-R were published in The Lancet Oncology.[1]

"While we are disappointed by this opinion, we will evaluate feedback received from the CHMP in order to determine next steps for quizartinib for the treatment of patients with relapsed/refractory FLT3-ITD AML in Europe," said Antoine Yver, MD, MSc, Executive Vice President and Global Head, Oncology Research and Development, Daiichi Sankyo. "Despite this setback, we continue to believe in the potential benefit of quizartinib for patients with FLT3-ITD AML and we look forward to the results of the global, pivotal phase 3 QuANTUM-First study evaluating quizartinib in combination with chemotherapy for patients with newly-diagnosed FLT3-ITD AML. We remain committed to bringing quizartinib forward as a potential treatment option for this aggressive and difficult-to-treat subtype of AML in the U.S., Europe and other parts of the world."

About Quizartinib
Quizartinib, an oral FLT3 inhibitor, is the lead product in the AML Franchise of Daiichi Sankyo.
Quizartinib currently is approved for use in Japan under the brand name VANFLYTA for the treatment of adult patients with relapsed/refractory FLT3-ITD AML, as detected by an approved test. It was launched in Japan on October 10, 2019.

Enrollment into QuANTUM-First, a global, pivotal phase 3 study evaluating quizartinib in combination with standard chemotherapy in newly diagnosed FLT3-ITD AML, was recently completed.

Other ongoing studies include phase 1/2 development for pediatric and young adult relapsed/refractory FLT3-ITD AML in North America and Europe; and phase 1 development in combination with milademetan, an investigational MDM2 inhibitor, for relapsed/refractory FLT3-ITD AML and newly-diagnosed FLT3-ITD AML unfit for intensive chemotherapy in the U.S. Milademetan is an investigational agent that has not been approved for any indication in any country. Safety and efficacy have not been established.

About FLT3-ITD AML
AML is an aggressive blood and bone marrow cancer that causes uncontrolled growth and accumulation of malignant white blood cells that fail to function normally and interfere with the production of normal blood cells.[2] In the EU, there are approximately 18,000 new cases of AML each year,[3] and their five-year survival rate is less than 30 percent.[4]

FLT3 gene mutations are one of the most common genetic abnormalities in AML.[5] FLT3-ITD is the most common FLT3 mutation, affecting approximately one in four patients with AML.[6] FLT3-ITD is a driver mutation that presents with high leukemic burden, a poor prognosis and a significant impact on disease management for patients with AML.[7] Patients with FLT3-ITD AML have a worse overall prognosis, including an increased incidence of relapse, an increased risk of death following relapse and a higher likelihood of relapse following hematopoietic stem cell transplantation, as compared to those without this mutation.[8],[9]

About Daiichi Sankyo Cancer Enterprise
The mission of Daiichi Sankyo Cancer Enterprise is to leverage our world-class, innovative science and push beyond traditional thinking to create meaningful treatments for patients with cancer. We are dedicated to transforming science into value for patients, and this sense of obligation informs everything we do. Anchored by three pillars, including our investigational Antibody Drug Conjugate Franchise, Acute Myeloid Leukemia Franchise and Breakthrough Science, we aim to deliver seven distinct new molecular entities over eight years during 2018 to 2025. Our powerful research engines include two laboratories for biologic/immuno-oncology and small molecules in Japan, and Plexxikon Inc., our small molecule structure-guided R&D center in Berkeley, CA. For more information, please visit: www.DSCancerEnterprise.com.

Castle Biosciences Presents Data at the ASDP 56th Annual Meeting Supporting DecisionDx-Melanoma Test’s Ability to Identify T1 Melanoma Patients at Low Risk for a Positive Sentinel Lymph Node

On October 18, 2019 Castle Biosciences, Inc. (Nasdaq: CSTL), a skin cancer diagnostics company providing personalized genomic information to improve cancer treatment decisions, reported the presentation of data supporting the ability of the DecisionDx-Melanoma gene expression profile (GEP) test to identify T1 (tumor depth of 1 mm or less) melanoma patients at low risk for a positive sentinel lymph node (SLN) at the American Society of Dermatopathology (ASDP) 56th Annual Meeting in San Diego from October 17-20 (Press release, Castle Biosciences, OCT 18, 2019, View Source [SID1234542365]).

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 study titled, "Identification of T1 melanoma patients at low risk for a positive sentinel lymph node (SLN) using a 31-gene expression profile (31-GEP)" (Abstract #262), will be presented as a poster at the meeting.

Study Background:

Current guidelines recommend that clinicians discuss the sentinel lymph node biopsy (SLNB) procedure with patients who have greater than 5% likelihood of SLN positivity; the guidelines do not recommend the procedure for patients with less than 5% likelihood of SLN positivity. Patients with T1 melanoma tumors have a 5.4% risk of SLN positivity overall.
The DecisionDx-Melanoma test is a prognostic test for cutaneous melanoma that predicts 5-year risk of metastasis as low risk (Class 1, 1A lowest risk) or high risk (Class 2, 2B highest risk).
The DecisionDx-Melanoma test has been previously validated to guide SLNB decisions, as patients with T1-T2 melanoma (tumor depth of 2 mm or less) with a Class 1A result had very low rates of SLN positivity.
This study was designed to evaluate the ability of the DecisionDx-Melanoma test to identify T1 melanoma patients with low risk for a positive SLN, using the combination of the previously published cohort with a novel cohort, totaling 910 consecutively tested T1 melanoma patients collected prospectively or retrospectively under IRB-approved protocols.
Key Findings:

For patients with T1 tumors of any age and a Class 1A test result, SLN positivity was 3.5%, significantly less than patients with a Class 1B-2B result (p=0.0005), and below the 5% threshold at which guidelines do not recommend the procedure.
For patients with T1 tumors 55 years of age or older and a Class 1A test result, SLN positivity was 2.3%, significantly less than patients with a Class 1B-2B result (p=0.002), and below the 5% threshold at which guidelines do not recommend the procedure.
Use of the DecisionDx-Melanoma test to guide SLNB decisions in patients with T1 melanoma could reduce SLNB surgical procedures by 80% in patients 55 years of age or older, as 272 of 339 patients who underwent this procedure had a Class 1A result.
Class 1A patients with T1 melanoma had recurrence free survival of 96.9% and distant metastasis-free survival of 97.3% on a retrospective dataset of 345 T1 patients with long-term follow-up, supporting that this population can safely avoid the SLNB surgical procedure.
"The study results show that use of the DecisionDx-Melanoma test can identify T1 patients who are at low risk for a positive sentinel lymph node and can guide sentinel lymph node biopsy discussions," said Federico A. Monzon, M.D., FCAP, chief medical officer at Castle Biosciences. "Furthermore, the identification of patients who are unlikely to benefit from sentinel lymph node biopsy can reduce unnecessary surgical procedures and have a positive impact on the allocation of healthcare resources."

About DecisionDx-Melanoma

DecisionDx-Melanoma is a gene expression profile test that uses an individual patient’s tumor biology to predict individual risk of cutaneous melanoma metastasis or recurrence, as well as sentinel lymph node positivity, independent of traditional staging factors, and has been studied in more than 3,900 patient samples. Using tissue from the primary melanoma, the test measures the expression of 31 genes. The test has been validated in four archival risk of recurrence studies of 901 patients and five prospective risk of recurrence studies including more than 780 patients. Prediction of the likelihood of sentinel lymph node positivity has also been validated in three prospective multicenter study cohorts that included more than 2,000 patients. Impact on patient management plans for one of every two patients tested has been demonstrated in four multicenter and single-center studies including more than 560 patients. The consistent performance and accuracy demonstrated in these studies provides confidence in disease management plans that incorporate DecisionDx-Melanoma test results.