Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Kenneth F. Grossmann is active.

Publication


Featured researches published by Kenneth F. Grossmann.


The New England Journal of Medicine | 2015

Combined nivolumab and ipilimumab or monotherapy in untreated Melanoma

Abstr Act; James Larkin; V. Chiarion Sileni; Rene Gonzalez; Dirk Schadendorf; Reinhard Dummer; Michael Smylie; Piotr Rutkowski; Andrew F. Hill; John Wagstaff; Michele Maio; Kenneth F. Grossmann; Mario Sznol; B. Dréno; Lars Bastholt; Arvin Yang; C. Horak; F. S. Hodi; Jedd D. Wolchok

BACKGROUND Nivolumab (a programmed death 1 [PD-1] checkpoint inhibitor) and ipilimumab (a cytotoxic T-lymphocyte-associated antigen 4 [CTLA-4] checkpoint inhibitor) have been shown to have complementary activity in metastatic melanoma. In this randomized, double-blind, phase 3 study, nivolumab alone or nivolumab plus ipilimumab was compared with ipilimumab alone in patients with metastatic melanoma. METHODS We assigned, in a 1:1:1 ratio, 945 previously untreated patients with unresectable stage III or IV melanoma to nivolumab alone, nivolumab plus ipilimumab, or ipilimumab alone. Progression-free survival and overall survival were coprimary end points. Results regarding progression-free survival are presented here. RESULTS The median progression-free survival was 11.5 months (95% confidence interval [CI], 8.9 to 16.7) with nivolumab plus ipilimumab, as compared with 2.9 months (95% CI, 2.8 to 3.4) with ipilimumab (hazard ratio for death or disease progression, 0.42; 99.5% CI, 0.31 to 0.57; P<0.001), and 6.9 months (95% CI, 4.3 to 9.5) with nivolumab (hazard ratio for the comparison with ipilimumab, 0.57; 99.5% CI, 0.43 to 0.76; P<0.001). In patients with tumors positive for the PD-1 ligand (PD-L1), the median progression-free survival was 14.0 months in the nivolumab-plus-ipilimumab group and in the nivolumab group, but in patients with PD-L1-negative tumors, progression-free survival was longer with the combination therapy than with nivolumab alone (11.2 months [95% CI, 8.0 to not reached] vs. 5.3 months [95% CI, 2.8 to 7.1]). Treatment-related adverse events of grade 3 or 4 occurred in 16.3% of the patients in the nivolumab group, 55.0% of those in the nivolumab-plus-ipilimumab group, and 27.3% of those in the ipilimumab group. CONCLUSIONS Among previously untreated patients with metastatic melanoma, nivolumab alone or combined with ipilimumab resulted in significantly longer progression-free survival than ipilimumab alone. In patients with PD-L1-negative tumors, the combination of PD-1 and CTLA-4 blockade was more effective than either agent alone. (Funded by Bristol-Myers Squibb; CheckMate 067 ClinicalTrials.gov number, NCT01844505.).


The New England Journal of Medicine | 2015

Nivolumab and Ipilimumab versus Ipilimumab in Untreated Melanoma

Michael A. Postow; Jason Chesney; Anna C. Pavlick; Caroline Robert; Kenneth F. Grossmann; David F. McDermott; Gerald P. Linette; Nicolas Meyer; Jeffrey K. Giguere; Sanjiv S. Agarwala; Montaser Shaheen; Marc S. Ernstoff; David R. Minor; April K. Salama; Matthew H. Taylor; Patrick A. Ott; Linda Rollin; Christine Horak; Paul Gagnier; Jedd D. Wolchok; F. Stephen Hodi

BACKGROUND In a phase 1 dose-escalation study, combined inhibition of T-cell checkpoint pathways by nivolumab and ipilimumab was associated with a high rate of objective response, including complete responses, among patients with advanced melanoma. METHODS In this double-blind study involving 142 patients with metastatic melanoma who had not previously received treatment, we randomly assigned patients in a 2:1 ratio to receive ipilimumab (3 mg per kilogram of body weight) combined with either nivolumab (1 mg per kilogram) or placebo once every 3 weeks for four doses, followed by nivolumab (3 mg per kilogram) or placebo every 2 weeks until the occurrence of disease progression or unacceptable toxic effects. The primary end point was the rate of investigator-assessed, confirmed objective response among patients with BRAF V600 wild-type tumors. RESULTS Among patients with BRAF wild-type tumors, the rate of confirmed objective response was 61% (44 of 72 patients) in the group that received both ipilimumab and nivolumab (combination group) versus 11% (4 of 37 patients) in the group that received ipilimumab and placebo (ipilimumab-monotherapy group) (P<0.001), with complete responses reported in 16 patients (22%) in the combination group and no patients in the ipilimumab-monotherapy group. The median duration of response was not reached in either group. The median progression-free survival was not reached with the combination therapy and was 4.4 months with ipilimumab monotherapy (hazard ratio associated with combination therapy as compared with ipilimumab monotherapy for disease progression or death, 0.40; 95% confidence interval, 0.23 to 0.68; P<0.001). Similar results for response rate and progression-free survival were observed in 33 patients with BRAF mutation-positive tumors. Drug-related adverse events of grade 3 or 4 were reported in 54% of the patients who received the combination therapy as compared with 24% of the patients who received ipilimumab monotherapy. Select adverse events with potential immunologic causes were consistent with those in a phase 1 study, and most of these events resolved with immune-modulating medication. CONCLUSIONS The objective-response rate and the progression-free survival among patients with advanced melanoma who had not previously received treatment were significantly greater with nivolumab combined with ipilimumab than with ipilimumab monotherapy. Combination therapy had an acceptable safety profile. (Funded by Bristol-Myers Squibb; ClinicalTrials.gov number, NCT01927419.).


Lancet Oncology | 2015

Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial

Jeffrey S. Weber; Sandra P. D'Angelo; David R. Minor; F. Stephen Hodi; Ralf Gutzmer; Bart Neyns; Christoph Hoeller; Nikhil I. Khushalani; Wilson H. Miller; Christopher D. Lao; Gerald P. Linette; Luc Thomas; Paul Lorigan; Kenneth F. Grossmann; Jessica C. Hassel; Michele Maio; Mario Sznol; Paolo Antonio Ascierto; Peter Mohr; Bartosz Chmielowski; Alan H. Bryce; Inge Marie Svane; Jean Jacques Grob; Angela M. Krackhardt; Christine Horak; Alexandre Lambert; Arvin Yang; James Larkin

BACKGROUND Nivolumab, a fully human IgG4 PD-1 immune checkpoint inhibitor antibody, can result in durable responses in patients with melanoma who have progressed after ipilimumab and BRAF inhibitors. We assessed the efficacy and safety of nivolumab compared with investigators choice of chemotherapy (ICC) as a second-line or later-line treatment in patients with advanced melanoma. METHODS In this randomised, controlled, open-label, phase 3 trial, we recruited patients at 90 sites in 14 countries. Eligible patients were 18 years or older, had unresectable or metastatic melanoma, and progressed after ipilimumab, or ipilimumab and a BRAF inhibitor if they were BRAF(V 600) mutation-positive. Participating investigators randomly assigned (with an interactive voice response system) patients 2:1 to receive an intravenous infusion of nivolumab 3 mg/kg every 2 weeks or ICC (dacarbazine 1000 mg/m(2) every 3 weeks or paclitaxel 175 mg/m(2) combined with carboplatin area under the curve 6 every 3 weeks) until progression or unacceptable toxic effects. We stratified randomisation by BRAF mutation status, tumour expression of PD-L1, and previous best overall response to ipilimumab. We used permuted blocks (block size of six) within each stratum. Primary endpoints were the proportion of patients who had an objective response and overall survival. Treatment was given open-label, but those doing tumour assessments were masked to treatment assignment. We assessed objective responses per-protocol after 120 patients had been treated with nivolumab and had a minimum follow-up of 24 weeks, and safety in all patients who had had at least one dose of treatment. The trial is closed and this is the first interim analysis, reporting the objective response primary endpoint. This study is registered with ClinicalTrials.gov, number NCT01721746. FINDINGS Between Dec 21, 2012, and Jan 10, 2014, we screened 631 patients, randomly allocating 272 patients to nivolumab and 133 to ICC. Confirmed objective responses were reported in 38 (31·7%, 95% CI 23·5-40·8) of the first 120 patients in the nivolumab group versus five (10·6%, 3·5-23·1) of 47 patients in the ICC group. Grade 3-4 adverse events related to nivolumab included increased lipase (three [1%] of 268 patients), increased alanine aminotransferase, anaemia, and fatigue (two [1%] each); for ICC, these included neutropenia (14 [14%] of 102), thrombocytopenia (six [6%]), and anaemia (five [5%]). We noted grade 3-4 drug-related serious adverse events in 12 (5%) nivolumab-treated patients and nine (9%) patients in the ICC group. No treatment-related deaths occurred. INTERPRETATION Nivolumab led to a greater proportion of patients achieving an objective response and fewer toxic effects than with alternative available chemotherapy regimens for patients with advanced melanoma that has progressed after ipilimumab or ipilimumab and a BRAF inhibitor. Nivolumab represents a new treatment option with clinically meaningful durable objective responses in a population of high unmet need. FUNDING Bristol-Myers Squibb.


Lancet Oncology | 2016

Combined nivolumab and ipilimumab versus ipilimumab alone in patients with advanced melanoma: 2-year overall survival outcomes in a multicentre, randomised, controlled, phase 2 trial

F. Stephen Hodi; Jason Chesney; Anna C. Pavlick; Caroline Robert; Kenneth F. Grossmann; David F. McDermott; Gerald P. Linette; Nicolas Meyer; Jeffrey K. Giguere; Sanjiv S. Agarwala; Montaser Shaheen; Marc S. Ernstoff; David R. Minor; April K. Salama; Matthew H. Taylor; Patrick A. Ott; Christine Horak; Paul Gagnier; Joel Jiang; Jedd D. Wolchok; Michael A. Postow

BACKGROUND Results from phase 2 and 3 trials in patients with advanced melanoma have shown significant improvements in the proportion of patients achieving an objective response and prolonged progression-free survival with the combination of nivolumab (an anti-PD-1 antibody) plus ipilimumab (an anti-CTLA-4 antibody) compared with ipilimumab alone. We report 2-year overall survival data from a randomised controlled trial assessing this treatment in previously untreated advanced melanoma. METHODS In this multicentre, double-blind, randomised, controlled, phase 2 trial (CheckMate 069) we recruited patients from 19 specialist cancer centres in two countries (France and the USA). Eligible patients were aged 18 years or older with previously untreated, unresectable stage III or IV melanoma and an Eastern Cooperative Oncology Group performance status of 0 or 1. Patients were randomly assigned 2:1 to receive an intravenous infusion of nivolumab 1 mg/kg plus ipilimumab 3 mg/kg or ipilimumab 3 mg/kg plus placebo, every 3 weeks for four doses. Subsequently, patients assigned to nivolumab plus ipilimumab received nivolumab 3 mg/kg every 2 weeks until disease progression or unacceptable toxicity, whereas patients allocated to ipilimumab alone received placebo every 2 weeks during this phase. Randomisation was done via an interactive voice response system with a permuted block schedule (block size of six) and stratification by BRAF mutation status. The study funder, patients, investigators, and study site staff were masked to treatment assignment. The primary endpoint, which has been reported previously, was the proportion of patients with BRAFV600 wild-type melanoma achieving an investigator-assessed objective response. Overall survival was an exploratory endpoint and is reported in this Article. Efficacy analyses were done on the intention-to-treat population, whereas safety was assessed in all treated patients who received at least one dose of study drug. This study is registered with ClinicalTrials.gov, number NCT01927419, and is ongoing but no longer enrolling patients. FINDINGS Between Sept 16, 2013, and Feb 6, 2014, we screened 179 patients and enrolled 142, randomly assigning 95 patients to nivolumab plus ipilimumab and 47 to ipilimumab alone. In each treatment group, one patient no longer met the study criteria following randomisation and thus did not receive study drug. At a median follow-up of 24·5 months (IQR 9·1-25·7), 2-year overall survival was 63·8% (95% CI 53·3-72·6) for those assigned to nivolumab plus ipilimumab and 53·6% (95% CI 38·1-66·8) for those assigned to ipilimumab alone; median overall survival had not been reached in either group (hazard ratio 0·74, 95% CI 0·43-1·26; p=0·26). Treatment-related grade 3-4 adverse events were reported in 51 (54%) of 94 patients who received nivolumab plus ipilimumab compared with nine (20%) of 46 patients who received ipilimumab alone. The most common treatment-related grade 3-4 adverse events were colitis (12 [13%] of 94 patients) and increased alanine aminotransferase (ten [11%]) in the combination group and diarrhoea (five [11%] of 46 patients) and hypophysitis (two [4%]) in the ipilimumab alone group. Serious grade 3-4 treatment-related adverse events were reported in 34 (36%) of 94 patients who received nivolumab plus ipilimumab (including colitis in ten [11%] of 94 patients, and diarrhoea in five [5%]) compared with four (9%) of 46 patients who received ipilimumab alone (including diarrhoea in two [4%] of 46 patients, colitis in one [2%], and hypophysitis in one [2%]). No new types of treatment-related adverse events or treatment-related deaths occurred in this updated analysis. INTERPRETATION Although follow-up of the patients in this study is ongoing, the results of this analysis suggest that the combination of first-line nivolumab plus ipilimumab might lead to improved outcomes compared with first-line ipilimumab alone in patients with advanced melanoma. The results suggest encouraging survival outcomes with immunotherapy in this population of patients. FUNDING Bristol-Myers Squibb.


Science Signaling | 2013

The Small GTPase ARF6 Stimulates β-Catenin Transcriptional Activity During WNT5A-Mediated Melanoma Invasion and Metastasis

Allie H. Grossmann; Jae Hyuk Yoo; James Clancy; Lise K. Sorensen; Alanna Sedgwick; Zongzhong Tong; Kirill Ostanin; Aaron Rogers; Kenneth F. Grossmann; Sheryl R. Tripp; Kirk R. Thomas; Crislyn D'Souza-Schorey; Shannon J. Odelberg; Dean Y. Li

WNT5A activates ARF6 to liberate β-catenin from adherens junctions, thereby increasing the amount that can mediate gene transcription. Releasing β-Catenin for Melanoma Metastasis Signaling through the Wnt pathway stimulates gene transcription mediated by β-catenin, and nuclear β-catenin is associated with a poor prognosis in cancer. The amount of β-catenin that is available to transcriptionally activate Wnt target genes may depend on the amount that is bound by cadherin in cell-cell adhesion structures called adherens junctions. Grossmann et al. found that WNT5A, which was produced by various melanoma cell lines, activated the small guanosine triphosphatase ARF6, triggered the release of β-catenin from N-cadherin, and increased the transcriptional activity of β-catenin. Pharmacological inhibition of ARF6 reduced β-catenin activity and invasive activity of melanoma cells in culture and decreased pulmonary metastasis in mice with tumors formed from melanoma cells. Thus, ARF6 may be a viable therapeutic target for reducing the invasiveness and metastasis of melanomas. β-Catenin has a dual function in cells: fortifying cadherin-based adhesion at the plasma membrane and activating transcription in the nucleus. We found that in melanoma cells, WNT5A stimulated the disruption of N-cadherin and β-catenin complexes by activating the guanosine triphosphatase adenosine diphosphate ribosylation factor 6 (ARF6). Binding of WNT5A to the Frizzled 4–LRP6 (low-density lipoprotein receptor–related protein 6) receptor complex activated ARF6, which liberated β-catenin from N-cadherin, thus increasing the pool of free β-catenin, enhancing β-catenin–mediated transcription, and stimulating invasion. In contrast to WNT5A, the guidance cue SLIT2 and its receptor ROBO1 inhibited ARF6 activation and, accordingly, stabilized the interaction of N-cadherin with β-catenin and reduced transcription and invasion. Thus, ARF6 integrated competing signals in melanoma cells, thereby enabling plasticity in the response to external cues. Moreover, small-molecule inhibition of ARF6 stabilized adherens junctions, blocked β-catenin signaling and invasiveness of melanoma cells in culture, and reduced spontaneous pulmonary metastasis in mice, suggesting that targeting ARF6 may provide a means of inhibiting WNT/β-catenin signaling in cancer.


Journal of Clinical Oncology | 2013

Severe Liver and Skin Toxicity After Radiation and Vemurafenib in Metastatic Melanoma

Christopher J. Anker; Antoni Ribas; Allie H. Grossmann; Xinjian Chen; Krishna K. Narra; Wallace Akerley; Robert H. I. Andtbacka; Robert Dirk Noyes; Dennis C. Shrieve; Kenneth F. Grossmann

Case Report A 15-year-old girl underwent wide local excision and sentinel lymphadenectomy of a thin, nonulcerated melanoma. One of 16 lymph nodes contained a focus of microscopic metastasis. Staging imaging showed no other disease. After the first of 12 planned months of adjuvant interferon alfa, the medication was discontinued early because of significant fatigue. New lung nodules were detected on surveillance computed tomography (CT) imaging 3 years after diagnosis, and a biopsy confirmed melanoma. Magnetic resonance imaging (MRI) of the brain also showed a new metastasis to the right parietal bone. The patient was started on high-dose interleukin-2. Follow-up MRI showed local progression of the skull lesion, so stereotactic radiation (RT) involving 25 Gy over five treatments was given after her second interleukin-2 cycle. Adverse effects of RT included alopecia and faint erythema in the radiated area. The patient developed back pain, and surveillance CT scans 2 weeks later showed new bone metastases in the axial skeleton, liver and spleen metastases, and progression in her lungs. BRAF mutation testing of a subcutaneous metastasis that was excised from the back showed a V600E mutation. The patient was enrolled onto a phase II study investigating vemurafenib in metastatic melanoma that was approved by the institutional review board of the University of California, Los Angeles. Her initial vemurafenib dose was 960 mg twice per day. Within 14 days, her performance status improved, with a substantial decrease in her spinal pain. No photosensitivity was observed. After 1 month, CT scans showed progression of bone metastases, but the disease in her liver, lungs, and spleen was either stable or decreased. After withholding vemurafenib for 4 days, 20 Gy of RT was administered over five fractions to the painful bone metastases. A posterior-anterior (PA) beam was used for T1 to T7 and T10 to L1, and her bilateral acetabula were treated with an AP/PA arrangement. Vemurafenib was restarted 2 days after the completion of RT. Two weeks after RT, the patient developed a tender, raised rash with well-delineated borders that matched her RT portals (Fig 1, portals are indicated in yellow; Fig 2, portals are indicated in cyan). Dry desquamation and then resolution of the skin changes occurred within 4 weeks. Imaging performed 3 weeks after RT showed overall stability of non-CNS disease, but 12 new brain metastases were detected. Although whole-brain therapy would be standard treatment in this scenario, because of significant concerns about skin toxicity, stereotactic radiosurgery (SRS) to each brain metastasis was recommended. Two weeks later the patient developed lower extremity weakness, and a lumbar spine MRI showed cauda equina compression at L4. She received 8 Gy of RT to L2 to L5 using a PA field, but vemurafenib was only withheld for 2 days because of the emergent nature of the treatment. Three days later, 20-Gy SRS was performed on each of the brain metastases. Vemurafenib was restarted 4 days after SRS, beginning at 480 mg for 3 days before moving to a full dose. Approximately 1 week after RT, she developed only mild erythema that matched the L2 to L5 portal. CT scans performed 10 weeks after the completion of her second course of RT showed interval pulmonary progression, with mixed responses elsewhere. Of concern was the development of innumerable, tightly packed, hypodense lesions in the liver that matched her previous RT portal (Fig 3A, pretreatment scan, black arrows indicate examples of liver metastases outside the RT portal; Fig 3B, posttreatment scan with RT isodose overlay [100 cGy 1 Gy]). Days later the patient developed severe chest discomfort and was admitted for pain control. The following 3 days she developed worsening abdominal pain and an acute drop in hematocrit. Interval accumulation of a large subcapsular hepatic hematoma and hemoperitoneum consistent with hepatic hemorrhage were detected on CT imaging (Fig 3C, white arrows). The patient died 2 days later. An autopsy showed an enlarged liver with multicystic change that was mostly limited to the central liver. Microscopically, these cysts were hemorrhagic and lined with melanoma cells (Fig 4A, 20 magnification of subcapsular cyst lined by melanoma. Organizing clot was seen emerging through ruptured cysts, explaining the sudden drop in hematocrit. Fig 4B, 40 magnification of multiple cysts lined by melanoma [arrow]; Fig 4C, 400 magnification). The intervening liver parenchyma showed severe zone III necrosis and scattered venous thrombi that were consistent with radiation-induced liver toxicity. However, outside of the radiation field, the liver also showed zone III necrosis, although it was less severe and without venous thrombi. This was suggestive of an additional source of liver damage subsequent to the initial insult, such as global ischemia. The late-occurring ischemic injury was likely the combined result of blood loss from a ruptured hematoma found at autopsy and multiorgan failure near the time of death.


Journal of Clinical Oncology | 2017

Efficacy and Safety Outcomes in Patients With Advanced Melanoma Who Discontinued Treatment With Nivolumab and Ipilimumab Because of Adverse Events: A Pooled Analysis of Randomized Phase II and III Trials.

Dirk Schadendorf; Jedd D. Wolchok; F. Stephen Hodi; Vanna Chiarion-Sileni; Rene Gonzalez; Piotr Rutkowski; Jean Jacques Grob; C. Lance Cowey; Christopher D. Lao; Jason Chesney; Caroline Robert; Kenneth F. Grossmann; David F. McDermott; Dana Walker; Rafia Bhore; James Larkin; Michael A. Postow

Purpose Approximately 40% of patients with advanced melanoma who received nivolumab combined with ipilimumab in clinical trials discontinued treatment because of adverse events (AEs). We conducted a retrospective analysis to assess the efficacy and safety of nivolumab plus ipilimumab in patients who discontinued treatment because of AEs. Methods Data were pooled from phase II and III trials of patients who received nivolumab 1 mg/kg plus ipilimumab 3 mg/kg, every 3 weeks for four doses, followed by nivolumab monotherapy 3 mg/kg every 2 weeks (N = 409). Efficacy was assessed in all randomly assigned patients who discontinued because of AEs during the induction phase (n = 96) and in those who did not discontinue because of AEs (n = 233). Safety was assessed in treated patients who discontinued because of AEs (n = 176) at any time and in those who did not discontinue because of AEs (n = 231). Results At a minimum follow-up of 18 months, median progression-free survival was 8.4 months for patients who discontinued treatment because of AEs during the induction phase and 10.8 months for patients who did not discontinue because of AEs ( P = .97). Median overall survival had not been reached in either group ( P = .23). The objective response rate was 58.3% for patients who discontinued because of AEs during the induction phase and 50.2% for patients who did not discontinue. The vast majority of grade 3 or 4 AEs occurred during the induction phase, with most resolving after appropriate management. Conclusion Efficacy outcomes seemed similar between patients who discontinued nivolumab plus ipilimumab treatment because of AEs during the induction phase and those who did not discontinue because of AEs. Therefore, even after discontinuation, many patients may continue to derive benefit from combination therapy.


Journal of Clinical Medicine | 2015

Detection of Exosomal miRNAs in the Plasma of Melanoma Patients.

Susan R. Pfeffer; Kenneth F. Grossmann; Pamela B. Cassidy; Chuan He Yang; Meiyun Fan; Levy Kopelovich; Sancy A. Leachman; Lawrence M. Pfeffer

MicroRNAs (miRNAs) are a class of 22–25 nucleotide RNAs that control gene expression at the post-transcriptional level. MiRNAs have potential as cancer biomarkers. Melanoma is a highly aggressive form of skin cancer accounting for almost 4% of cancers among men and women, and ~80% of skin cancer-related deaths in the US. In the present study we analyzed plasma-derived exosomal miRNAs from clinically affected and unaffected familial melanoma patients (CDKN2A/p16 gene carriers) and compared them with affected (nonfamilial melanoma) and unaffected control subjects in order to identify novel risk biomarkers for melanoma. Intact miRNAs can be isolated from the circulation because of their presence in exosomes. A number of differentially regulated miRNAs identified by NanoString human V2 miRNA array were validated by quantitative PCR. Significantly, miR-17, miR-19a, miR-21, miR-126, and miR-149 were expressed at higher levels in patients with metastatic sporadic melanoma as compared with familial melanoma patients or unaffected control subjects. Surprisingly, no substantial differences in miRNA expression were detected between familial melanoma patients (all inclusive) and unaffected control subjects. The miRNAs differentially expressed in the different patient cohorts, especially in patients with metastatic melanoma, may play important roles in tumor progression and metastasis, and may be used as predictive biomarkers to monitor remission as well as relapse following therapeutic intervention.


Journal of Clinical Oncology | 2017

Overall Survival in Patients With Advanced Melanoma Who Received Nivolumab Versus Investigator’s Choice Chemotherapy in CheckMate 037: A Randomized, Controlled, Open-Label Phase III Trial

James Larkin; David R. Minor; Sandra P. D'Angelo; Bart Neyns; Michael Smylie; Wilson H. Miller; Ralf Gutzmer; Gerald P. Linette; Bartosz Chmielowski; Christopher D. Lao; Paul Lorigan; Kenneth F. Grossmann; Jessica C. Hassel; Mario Sznol; Adil Daud; Jeffrey A. Sosman; Nikhil I. Khushalani; Dirk Schadendorf; Christoph Hoeller; Dana Walker; George Kong; Christine Horak; Jeffrey S. Weber

Purpose Until recently, limited options existed for patients with advanced melanoma who experienced disease progression while receiving treatment with ipilimumab. Here, we report the coprimary overall survival (OS) end point of CheckMate 037, which has previously shown that nivolumab resulted in more patients achieving an objective response compared with chemotherapy regimens in ipilimumab-refractory patients with advanced melanoma. Patients and Methods Patients were stratified by programmed death-ligand 1 expression, BRAF status, and best prior cytotoxic T-lymphocyte antigen-4 therapy response, then randomly assigned 2:1 to nivolumab 3 mg/kg intravenously every 2 weeks or investigators choice chemotherapy (ICC; dacarbazine 1,000 mg/m2 every 3 weeks or carboplatin area under the curve 6 plus paclitaxel 175 mg/m2 every 3 weeks). Patients were treated until they experienced progression or unacceptable toxicity, with follow-up of approximately 2 years. Results Two hundred seventy-two patients were randomly assigned to nivolumab (99% treated) and 133 to ICC (77% treated). More nivolumab-treated patients had brain metastases (20% v 14%) and increased lactate dehydrogenase levels (52% v 38%) at baseline; 41% of patients treated with ICC versus 11% of patients treated with nivolumab received anti-programmed death 1 agents after randomly assigned therapy. Median OS was 16 months for nivolumab versus 14 months for ICC (hazard ratio, 0.95; 95.54% CI, 0.73 to 1.24); median progression-free survival was 3.1 months versus 3.7 months, respectively (hazard ratio, 1.0; 95.1% CI, 0.78 to 1.436). Overall response rate (27% v 10%) and median duration of response (32 months v 13 months) were notably higher for nivolumab versus ICC. Fewer grade 3 and 4 treatment-related adverse events were observed in patients on nivolumab (14% v 34%). Conclusion Nivolumab demonstrated higher, more durable responses but no difference in survival compared with ICC. OS should be interpreted with caution as it was likely impacted by an increased dropout rate before treatment, which led to crossover therapy in the ICC group, and by an increased proportion of patients in the nivolumab group with poor prognostic factors.


Journal of Neurosurgery | 2015

Local control after stereotactic radiosurgery for brain metastases in patients with melanoma with and without BRAF mutation and treatment

David Ly; Hilary P. Bagshaw; Christopher J. Anker; Kenneth F. Grossmann; Randy L. Jensen; Dennis C. Shrieve

OBJECT BRAF inhibitors improve progression-free and overall survival in patients with metastatic melanoma. Brain metastases are common, and stereotactic radiosurgery (SRS) has been used, resulting in excellent local control. Because BRAF inhibitors are associated with intracranial responses, the authors hypothesized that BRAF inhibitors would improve local control in patients with melanoma who are receiving SRS for brain metastases. METHODS The authors retrospectively identified patients with metastatic melanoma who had been tested for BRAF mutation and treated with SRS for brain metastases. Patients with previous resection, multiple brain metastases, or multiple courses of SRS were eligible. SRS was delivered in a single fraction to a median dose of 2000 cGy. Patients with a BRAF mutation were treated with a BRAF inhibitor on the basis of physician preference. RESULTS The authors identified 52 patients who were treated in 82 treatment sessions for 185 brain metastases and 13 tumor beds. At a median follow-up of 10.5 months, the 1-year local control rate was 69.2%. At 1 year, the local control rate for brain metastases in patients with BRAF mutation with BRAF treatment was 85.0%, and the local control rate for brain metastases in those without BRAF treatment was 51.5% (p = 0.0077). The rates of distant brain failure, freedom from whole-brain radiation, and overall survival were not different on the basis of BRAF mutation status or inhibitor therapy. The number of new intratumoral hemorrhages after SRS was increased significantly in patients with BRAF treatment. CONCLUSIONS Treatment with BRAF inhibitors was associated with improved local control after SRS in patients with melanoma and brain metastases. An increased number of intratumoral hemorrhages was associated with BRAF inhibitor therapy.

Collaboration


Dive into the Kenneth F. Grossmann's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Antoni Ribas

University of California

View shared research outputs
Top Co-Authors

Avatar

David R. Minor

California Pacific Medical Center

View shared research outputs
Top Co-Authors

Avatar

Gerald P. Linette

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Jedd D. Wolchok

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge