Network


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

Hotspot


Dive into the research topics where Dylan J. Martini is active.

Publication


Featured researches published by Dylan J. Martini.


Cancer immunology research | 2018

Durable clinical benefit in metastatic renal cell carcinoma patients who discontinue PD-1/PD-L1 therapy for immune-related adverse events (irAEs)

Dylan J. Martini; Lana Hamieh; Rana R. McKay; Lauren C. Harshman; Raphael Brandao; Craig Norton; John A. Steinharter; Katherine M. Krajewski; Xin Gao; Fabio A.B. Schutz; Bradley Alexander McGregor; Dominick Bossé; Aly-Khan A. Lalani; Guillermo Velasco; M. Dror Michaelson; David F. McDermott; Toni K. Choueiri

mRCC patients who had a clinical response to PD-1/PD-L1 blockade and discontinued treatment due to irAE continued to derive prolonged clinical benefit after treatment was stopped. Current clinical trials are exploring customized approaches to application of PD-1/PD-L1 blockade. The current standard of care for treatment of metastatic renal cell carcinoma (mRCC) patients is PD-1/PD-L1 inhibitors until progression or toxicity. Here, we characterize the clinical outcomes for 19 mRCC patients who experienced an initial clinical response (any degree of tumor shrinkage), but after immune-related adverse events (irAE) discontinued all systemic therapy. Clinical baseline characteristics, outcomes, and survival data were collected. The primary endpoint was time to progression from the date of treatment cessation (TTP). Most patients had clear cell histology and received anti–PD–1/PD-L1 therapy as second-line or later treatment. Median time on PD-1/PD-L1 therapy was 5.5 months (range, 0.7–46.5) and median TTP was 18.4 months (95% CI, 4.7–54.3) per Kaplan–Meier estimation. The irAEs included arthropathies, ophthalmopathies, myositis, pneumonitis, and diarrhea. We demonstrate that 68.4% of patients (n = 13) experienced durable clinical benefit off treatment (TTP of at least 6 months), with 36% (n = 7) of patients remaining off subsequent treatment for over a year after their last dose of anti–PD-1/PD-L1. Three patients with tumor growth found in a follow-up visit, underwent subsequent surgical intervention, and remain off systemic treatment. Nine patients (47.4%) have ongoing irAEs. Our results show that patients who benefitted clinically from anti–PD-1/PD-L1 therapy can experience sustained beneficial responses, not needing further therapies after the initial discontinuation of treatment due to irAEs. Investigation of biomarkers indicating sustained benefit to checkpoint blockers are needed. Cancer Immunol Res; 6(4); 402–8. ©2018 AACR.


Journal for ImmunoTherapy of Cancer | 2017

Response to single agent PD-1 inhibitor after progression on previous PD-1/PD-L1 inhibitors: a case series

Dylan J. Martini; Aly-Khan A. Lalani; Dominick Bossé; John A. Steinharter; Lauren C. Harshman; F. Stephen Hodi; Patrick A. Ott; Toni K. Choueiri

Background Monoclonal antibodies targeting the PD-1/PD-L1 axis have gained increasing attention across many solid tumors and hematologic malignancies due to their efficacy and favorable toxicity profile. With more than 1 agent now FDA-approved in a wide variety of tumor types, and with others in clinical trials, it is becoming more common that patients present to clinic for potential treatment with a second PD-1/PD-L1 inhibitor. Case presentation In this report, we present two patients with renal cell carcinoma and one with melanoma who received PD-1/PD-L1 inhibitors. Upon progression on their first-line PD-1/PD-L1 inhibitors, these patients received a different PD-1 inhibitor (nivolumab in all cases) and all had progressive disease as their best response to the subsequent PD-1 inhibitor. The reported clinical information focuses on the course of the disease and the responses to all treatment regimens. Conclusions Clinicians should refrain from using multiple PD-1/PD-L1 inhibitors sequentially outside of clinical trials until there is sufficient data to support this practice routinely. Prospective studies that allow prior treatment with PD-1/PD-L1 are needed to validate the efficacy and safety of these drugs in the second line or later setting. Furthermore, ongoing efforts that aim to identify mechanisms of resistance to immunotherapy will be informative and may ultimately assist physicians in select the optimal treatment following progression on PD-1/PD-L1 inhibitor.BackgroundMonoclonal antibodies targeting the PD-1/PD-L1 axis have gained increasing attention across many solid tumors and hematologic malignancies due to their efficacy and favorable toxicity profile. With more than 1 agent now FDA-approved in a wide variety of tumor types, and with others in clinical trials, it is becoming more common that patients present to clinic for potential treatment with a second PD-1/PD-L1 inhibitor.Case presentationIn this report, we present two patients with renal cell carcinoma and one with melanoma who received PD-1/PD-L1 inhibitors. Upon progression on their first-line PD-1/PD-L1 inhibitors, these patients received a different PD-1 inhibitor (nivolumab in all cases) and all had progressive disease as their best response to the subsequent PD-1 inhibitor. The reported clinical information focuses on the course of the disease and the responses to all treatment regimens.ConclusionsClinicians should refrain from using multiple PD-1/PD-L1 inhibitors sequentially outside of clinical trials until there is sufficient data to support this practice routinely. Prospective studies that allow prior treatment with PD-1/PD-L1 are needed to validate the efficacy and safety of these drugs in the second line or later setting. Furthermore, ongoing efforts that aim to identify mechanisms of resistance to immunotherapy will be informative and may ultimately assist physicians in select the optimal treatment following progression on PD-1/PD-L1 inhibitor.


Cancer immunology research | 2018

The Clinical Activity of PD-1/PD-L1 Inhibitors in Metastatic Non–Clear Cell Renal Cell Carcinoma

Rana R. McKay; Dominick Bossé; Wanling Xie; Stephanie A. Wankowicz; Abdallah Flaifel; Raphael Brandao; Aly-Khan A. Lalani; Dylan J. Martini; Xiao X. Wei; David A. Braun; Eliezer M. Van Allen; Daniel Castellano; Guillermo Velasco; J. Connor Wells; Daniel Y.C. Heng; Andre Poisl Fay; Fabio A.B. Schutz; JoAnn Hsu; Sumanta K. Pal; James J. Hsieh; Lauren C. Harshman; Sabina Signoretti; Robert J. Motzer; Darren R. Feldman; Toni K. Choueiri

A multicenter pooled analysis revealed modest antitumor activity of PD-1/PD-L1 inhibitors in patients with rare kidney cancer subtypes. Programmed death 1 (PD-1) and PD ligand 1 (PD-L1) inhibitors have shown activity in metastatic clear cell renal cell carcinoma (ccRCC). Data on the activity of these agents in patients with non–clear cell RCC (nccRCC) or patients with sarcomatoid/rhabdoid differentiation are limited. In this multicenter analysis, we explored the efficacy of PD-1/PD-L1 inhibitors in patients with nccRCC or sarcomatoid/rhabdoid differentiation. Baseline and follow-up demographic, clinical, treatment, and radiographic data were collected. The primary endpoint was objective response rate. Secondary endpoints include time-to-treatment failure (TTF), overall survival (OS), and biomarker correlates. Forty-three patients were included: papillary (n = 14; 33%), chromophobe (n = 10; 23%), unclassified (n = 9; 21%), translocation (n = 3; 7%), and ccRCC with sarcomatoid differentiation (n = 7, 16%). Of those 43 patients, 11 patients (26%) had sarcomatoid and/or rhabdoid differentiation (n = 7 with ccRCC; n = 4 nccRCC). Overall, 8 patients (19%) objectively responded, including 4 patients (13%) who received PD-1/PD-L1 monotherapy. Responses were observed in patients with ccRCC with sarcomatoid and/or rhabdoid differentiation (n = 3/7, 43%), translocation RCC (n = 1/3, 33%), and papillary RCC (n = 4/14, 29%). The median TTF was 4.0 months [95% confidence interval (CI), 2.8–5.5] and median OS was 12.9 months (95% CI, 7.4–not reached). No specific genomic alteration was associated with clinical benefit. Modest antitumor activity for PD-1/PD-L1–blocking agents was observed in some patients with nccRCC. Further prospective studies are warranted to investigate the efficacy of PD-1/PD-L1 blockade in this heterogeneous patient population. Cancer Immunol Res; 6(7); 758–65. ©2018 AACR.


Cancer immunology research | 2017

Abstract A18: Genomic and neoantigen evolution and resistance to immune checkpoint blockade in metastatic renal cell carcinoma

Diana Miao; Guillermo Velasco; Craig Norton; Dylan J. Martini; Stephanie A. Mullane; Raphael Brandao Moreira; Sabina Signoretti; Kwok-Kin Wong; Toni K. Choueiri; Eliezer M. Van Allen

Blockade of the PD1/PD-L1 T cell immune checkpoint yields responses and survival benefits exceeding mTOR blockade alone in metastatic renal cell carcinoma (mRCC). However, predictors of response to anti-PD1/PD-L1 therapy are mostly unknown, and pathways of acquired resistance to immune checkpoint therapy are poorly understood. In this study, we undertook whole exome sequencing and neoantigen analysis of matched “trios” (primary tumor resection, immune-checkpoint-refractory metastasis, and germline tissue) from 3 patients treated with anti-PD1 or anti-PD-L1 therapy for mRCC. Patients RCC-001 and RC-002 received anti-PD1 therapy after VEGF-targeted therapy and patient RCC-003 received anti-PD-L1 first-line for mRCC. All 3 patients experienced tumor regression on immune checkpoint therapy, with treatment durations of 10 months, 2 years, and 3 months, respectively. Progressive disease samples were sequenced for analysis of genomic mechanisms of treatment resistance. Whole transcriptome sequencing was obtained on primary and metastatic tumors from RCC-003. Nonsynonymous mutation load was moderate in all samples (range: 33-46 primary, 36-67 resistant). Alterations in VHL were seen in RCC-001 and RCC-002 before and after treatment. RCC-003 had a frameshift deletion in the tumor suppressor NF2 and a nonsense mutation in the MHCI antigen-processing protein TAP1 in both primary and refractory tumors. Resistance-associated mutations in RCC-002 included a frameshift deletion in MR1, involved in MHC I antigen presentation, and missense mutations in TJP1, implicated in JAK/STAT signaling, and PIAS2, implicated in STAT2 and PTEN regulation. JAK2 amplifications have been found to upregulate PD-L1 expression (Green et al. 2010, Blood), and PTEN loss mediates resistance to T cell-mediated immmunotherapy in vitro (Peng et al. 2016, Cancer Discovery). None of the primary tumors harbored alterations in microsatellite-instability-associated genes, and loss of β2 microglobulin was not observed in any sample. Despite moderate mutational loads, each sample harbored a sizeable number of neoantigens (range: 24-76 primary, 50-104 resistant). Across the 6 samples, 28 to 69% of nonsynonymous mutations were predicted to yield at least one neoantigen, with one alteration yielding a maximum of 12 unique neoantigens. Up to 60% of neoantigens observed in the primary tumor were not seen in the resistant setting (range: 20.8-59.2%). Whole transcriptome sequencing in RCC-003 showed 18/24 predicted neoantigens were expressed in the primary tumor and 24/50 in the refractory sample. 13 neoantigens were shared between the two samples, while 5 were unique to the pre-treatment tumor. Integrated whole exome and whole transcriptome sequencing with matched germline profiling identified 5 neoantigens in RCC-003 that were found exclusively in the primary tumor (lost in the treatment-resistant tumor). This could represent immune evasion via deletion of immunogenic mutations, cytotoxic killing of immunogenic tumor clones, or intrinsic resistance to immune checkpoint therapy in heterogenous tumors. Ongoing in vitro analysis of reactivity of predicted neoantigens from all 3 samples with patient-derived T cells will further clarify the biological significance these putative tumor antigens. Citation Format: Diana Miao, Guillermo De Velasco, Dennis Adeegbe, Craig Norton, Dylan Martini, Stephanie Mullane, Raphael Moreira, Sabina Signoretti, Kwok-Kin Wong, Toni Choueiri, Eliezer Van Allen. Genomic and neoantigen evolution and resistance to immune checkpoint blockade in metastatic renal cell carcinoma. [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2016 Oct 20-23; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2017;5(3 Suppl):Abstract nr A18.


Cancer | 2018

The prognostic and predictive impact of inflammatory biomarkers in patients who have advanced-stage cancer treated with immunotherapy: Inflammatory Biomarkers in Immunotherapy

Mehmet Asim Bilen; Dylan J. Martini; Yuan Liu; Colleen Lewis; Hannah Collins; Julie M. Shabto; Mehmet Akce; Haydn T. Kissick; Bradley C. Carthon; Walid Labib Shaib; Olatunji B. Alese; Rathi N. Pillai; Conor E. Steuer; Christina Wu; David H. Lawson; Ragini R. Kudchadkar; Bassel F. El-Rayes; Viraj A. Master; Suresh S. Ramalingam; Taofeek K. Owonikoko; R. Donald Harvey

Optimal prognostic and predictive biomarkers for patients with advanced‐stage cancer patients who received immunotherapy (IO) are lacking. Inflammatory markers, such as the neutrophil‐to‐lymphocyte ratio (NLR), the monocyte‐to‐lymphocyte ratio (MLR), and the platelet‐to‐lymphocyte ratio (PLR), are readily available. The authors investigated the association between these markers and clinical outcomes of patients with advanced‐stage cancer who received IO.


Science | 2018

Genomic correlates of response to immune checkpoint therapies in clear cell renal cell carcinoma

Diana Miao; Claire Margolis; Wenhua Gao; Martin H. Voss; Wei Li; Dylan J. Martini; Craig Norton; Dominick Bossé; Stephanie M. Wankowicz; Dana Cullen; Christine Horak; Megan Wind-Rotolo; Adam Tracy; Marios Giannakis; F.S. Hodi; Charles G. Drake; Mark W. Ball; Mohamad E. Allaf; Alexandra Snyder; Matthew D. Hellmann; Thai H. Ho; Robert J. Motzer; Sabina Signoretti; William G. Kaelin; Toni K. Choueiri; Eliezer M. Van Allen


Journal for ImmunoTherapy of Cancer | 2018

Change in Neutrophil-to-lymphocyte ratio (NLR) in response to immune checkpoint blockade for metastatic renal cell carcinoma

Aly-Khan A. Lalani; Wanling Xie; Dylan J. Martini; John A. Steinharter; Craig Norton; Katherine M. Krajewski; Audrey Duquette; Dominick Bossé; Joaquim Bellmunt; Eliezer M. Van Allen; Bradley Alexander McGregor; Chad J. Creighton; Lauren C. Harshman; Toni K. Choueiri


Journal of Clinical Oncology | 2018

Antibiotic use and outcomes with systemic therapy in metastatic renal cell carcinoma (mRCC).

Aly-Khan A. Lalani; Wanling Xie; Xun Lin; John A. Steinharter; Dylan J. Martini; Audrey Duquette; Dominick Bossé; Rana R. McKay; Ronit Simantov; Xiao X. Wei; Bradley Alexander McGregor; Lauren C. Harshman; Toni K. Choueiri


Journal of Clinical Oncology | 2017

Outcomes of PD-1/PD-L1 responders who discontinue therapy for immune-related adverse events (irAEs): Results of a cohort of patients (pts) with metastatic renal cell carcinoma (mRCC).

Rana R. McKay; Dylan J. Martini; Raphael Brandao Moreira; Lana Hamieh; Craig Norton; Stephanie A. Mullane; Meghara K. Walsh; Dror Michaelson; David F. McDermott; Eliezer VanAllen; Lauren C. Harshman; Toni K. Choueiri


Journal of Clinical Oncology | 2017

Loss-of-function of PBRM1 to predict response to anti-PD-1/PD-L1 therapy in metastatic renal cell carcinoma.

Diana Miao; Claire Margolis; Dylan J. Martini; Stephanie A. Mullane; Dana Cullen; Christine Horak; Megan Wind-Rotolo; Matthew D. Hellmann; Martin H. Voss; Robert J. Motzer; Craig Norton; Sabina Signoretti; Alexandra Snyder Charen; Eliezer M. Van Allen; Toni K. Choueiri

Collaboration


Dive into the Dylan J. Martini's collaboration.

Top Co-Authors

Avatar

Toni K. Choueiri

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge