Network


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

Hotspot


Dive into the research topics where Benjamin A. Weinberg is active.

Publication


Featured researches published by Benjamin A. Weinberg.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Abnormal lymphangiogenesis in idiopathic pulmonary fibrosis with insights into cellular and molecular mechanisms

Souheil El-Chemaly; Daniela Malide; Zudaire E; Yoshihiko Ikeda; Benjamin A. Weinberg; Gustavo Pacheco-Rodriguez; Ivan O. Rosas; Aparicio M; Ping Ren; Sandra D. MacDonald; Hai-Ping Wu; Steven D. Nathan; Cuttitta F; McCoy Jp; Bernadette R. Gochuico; Joel Moss

Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, debilitating respiratory disease whose pathogenesis is poorly understood. In IPF, the lung parenchyma undergoes extensive remodeling. We hypothesized that lymphangiogenesis is part of lung remodeling and sought to characterize pathways leading to lymphangiogenesis in IPF. We found that the diameter of lymphatic vessels in alveolar spaces in IPF lung tissue correlated with disease severity, suggesting that the alveolar microenvironment plays a role in the lymphangiogenic process. In bronchoalveolar lavage fluid (BALF) from subjects with IPF, we found short-fragment hyaluronic acid, which induced migration and proliferation of lymphatic endothelial cells (LECs), processes required for lymphatic vessel formation. To determine the origin of LECs in IPF, we isolated macrophages from the alveolar spaces; CD11b+ macrophages from subjects with IPF, but not those from healthy volunteers, formed lymphatic-like vessels in vitro. Our findings demonstrate that in the alveolar microenvironment of IPF, soluble factors such as short-fragment hyaluronic acid and cells such as CD11b+ macrophages contribute to lymphangiogenesis. These results improve our understanding of lymphangiogenesis and tissue remodeling in IPF and perhaps other fibrotic diseases as well.


Oncotarget | 2017

Comparative molecular analyses of left-sided colon, right-sided colon, and rectal cancers

Mohamed E. Salem; Benjamin A. Weinberg; Joanne Xiu; Wafik S. El-Deiry; Jimmy J. Hwang; Zoran Gatalica; Philip A. Philip; Anthony F. Shields; Heinz-Josef Lenz; John L. Marshall

Tumor sidedness has emerged as an important prognostic and predictive factor in the treatment of colorectal cancer. Recent studies demonstrate that patients with advanced right-sided colon cancers have a worse prognosis than those with left-sided colon or rectal cancers, and these patient subgroups respond differently to biological therapies. Historically, management of patients with metastatic colon and rectal cancers has been similar, and colon and rectal cancer patients have been grouped together in large clinical trials. Clearly, the differences in molecular biology among right-sided colon, left-sided colon, and rectal cancers should be further studied in order to account for disparities in clinical outcomes. We profiled 10,570 colorectal tumors (of which 2,413 were identified as arising from the left colon, right colon, or rectum) using next-generation sequencing, immunohistochemistry, chromogenic in-situ hybridization, and fragment analysis (Caris Life Sciences, Phoenix, AZ). Right-sided colon cancers had higher rates of microsatellite instability, more frequent aberrant activation of the EGFR pathway including higher BRAF and PIK3CA mutation rates, and increased mutational burden compared to left-sided colon and rectal cancers. Rectal cancers had higher rates of TOPO1 expression and Her2/neu amplification compared to both left- and right-sided colon cancers. Molecular variations among right-sided colon, left-sided colon, and rectal tumors may contribute to differences in clinical behavior. The site of tumor origin (left colon, right colon, or rectum) should certainly be considered when selecting treatment regimens and stratifying patients for future clinical trials.


Oncologist | 2018

Impact of Patient Age on Molecular Alterations of Left‐Sided Colorectal Tumors

Alberto Puccini; Heinz-Josef Lenz; John L. Marshall; David Arguello; Derek Raghavan; W. Michael Korn; Benjamin A. Weinberg; Kelsey Poorman; Arielle L. Heeke; Philip A. Philip; Anthony F. Shields; Richard M. Goldberg; Mohamed E. Salem

BACKGROUND The incidence of colorectal cancer (CRC) in younger patients is rising, mostly due to tumors in the descending colon and rectum. Therefore, we aimed to explore the molecular differences of left-sided CRC between younger (≤45 years) and older patients (≥65). SUBJECTS, MATERIALS, AND METHODS In total, 1,126 CRC tumor samples from the splenic flexure to (and including) the rectum were examined by next-generation sequencing (NGS), immunohistochemistry, and in situ hybridization. Microsatellite instability (MSI) and tumor mutational burden (TMB) were assessed by NGS. RESULTS Younger patients (n = 350), when compared with older patients (n = 776), showed higher mutation rates in genes associated with cancer-predisposing syndromes (e.g., Lynch syndrome), such as MSH6 (4.8% vs. 1.2%, p = .005), MSH2 (2.7% vs. 0.0%, p = .004), POLE (1.6% vs. 0.0%, p = .008), NF1 (5.9% vs. 0.5%, p < .001), SMAD4 (14.3% vs. 8.3%, p = .024), and BRCA2 (3.7% vs. 0.5%, p = .002). Genes involved in histone modification were also significantly more mutated: KDM5C (1.9% vs. 0%, p = .036), KMT2A (1.1% vs. 0%, p = .033), KMT2C (1.6% vs. 0%, p = .031), KMT2D (3.8% vs. 0.7%, p = .005), and SETD2 (3.2% vs. 0.9%, p = .039). Finally, TMB-high (9.7% vs. 2.8%, p < .001) and MSI-high (MSI-H; 8.1% vs. 1.9%, p = .009) were more frequent in younger patients. CONCLUSION Our findings highlight the importance of genetic counseling and screening in younger CRC patients. MSI-H and TMB-high tumors could benefit from immune-checkpoint inhibitors, now approved for the treatment of MSI-H/deficient mismatch repair metastatic CRC patients. Finally, histone modifiers could serve as a new promising therapeutic target. With confirmatory studies, these results may influence our approach to younger adults with CRC. IMPLICATIONS FOR PRACTICE The increasing rate of colorectal cancers (CRC), primarily distal tumors, among young adults poses a global health issue. This study investigates the molecular differences between younger (≤45 years old) and older (≥65) adults with left-sided CRCs. Younger patients more frequently harbor mutations in genes associated with cancer-predisposing syndromes. Higher rates of microsatellite instability-high and tumor mutational burden-high tumors occur in younger patients, who could benefit from immune-checkpoint inhibitors. Finally, histone modifiers are more frequently mutated in younger patients and could serve as a new promising therapeutic target. This study provides new insights into mutations that may guide development of novel tailored therapy in younger CRC patients.


Oncologist | 2018

Immuno‐Oncology Biomarkers for Gastric and Gastroesophageal Junction Adenocarcinoma: Why PD‐L1 Testing May Not Be Enough

Benjamin A. Weinberg; Joanne Xiu; Jimmy J. Hwang; Anthony F. Shields; Mohamed E. Salem; John L. Marshall

PURPOSE The treatment of patients with advanced gastric and gastroesophageal junction (G/GEJ) adenocarcinomas has been transformed by the U.S. Food and Drug Administration approval of pembrolizumab. Tumor and adjacent tissue must stain positively for the programmed cell death ligand 1 (PD-L1) protein by companion diagnostic testing. However, some patients with PD-L1-negative tumors also benefit from pembrolizumab. High microsatellite instability (MSI) and tumor mutational load (TML) are positive predictive biomarkers for immune checkpoint inhibition (ICI) in other tumors. We sought to identify more patients who could benefit from ICI using alternative PD-L1 thresholds, MSI, and TML. METHODS Tumor specimens underwent next-generation sequencing (NGS) and PD-L1 testing using immunohistochemistry. NGS was used to determine TML and MSI. RESULTS We profiled 581 G/GEJ adenocarcinoma specimens. PD-L1 staining was scored for intensity (0, none; 1+, weak; 2+, moderate; 3+, strong). Using 2+ staining at a 5% threshold, 9.3% of tumors were PD-L1 positive, and using 1+ staining at 1%, 16.2% were PD-L1 positive. 6.9% of tumors had high MSI. High TML (≥17 mutations per megabase) was seen in 6.9%, and medium TML (≥7) was seen in 56.5% of tumors. Thirty (5.2%) PD-L1-negative tumors at the 1+, 1% threshold had high TML or high MSI. Primary tumors had higher rates of high TML (8.8% vs. 3.9%; p = .0377) and high MSI (8.5% vs. 3.9%; p = .0471) than metastases. CONCLUSION PD-L1 testing alone fails to detect patients who may benefit from ICI. Lower PD-L1 thresholds and TML testing should be considered in future clinical trials. IMPLICATIONS FOR PRACTICE Pembrolizumab is approved by the U.S. Food and Drug Administration for patients with refractory gastric and gastroesophageal cancers if the tumor and adjacent tissue stain positively for the programmed cell death ligand 1 (PD-L1) protein by companion diagnostic testing. Tumor mutational load, microsatellite instability (MSI), and alternative PD-L1 testing thresholds may serve as predictive biomarkers for response to immune checkpoint inhibition, and standard PD-L1 testing will not identify all patients who may benefit from this therapy.


Archive | 2018

Vaccine Therapy in Pancreatic Cancer

Benjamin A. Weinberg; Michael J. Pishvaian

Pancreatic adenocarcinoma is an aggressive malignancy associated with poor overall survival. There is a dire need for novel targeted agents to add to the armamentarium of therapeutics for this disease. Vaccines represent an important method to train the body’s own immune system to attack pancreatic cancer cells. Despite a number of unique targets on pancreatic cancer cells, vaccines have been largely ineffective at prolonging survival in pancreatic cancer patients due to poor immunogenicity and failure to translate an immune response into a clinical response. Numerous attempts have been made to augment vaccine administration using different vaccine constructs, adjuvants, growth factors, and chemotherapy combinations, but durable clinical benefit remains elusive. New vaccine combinations with checkpoint inhibitors offer hope of unleashing the full potential of vaccine therapy. We highlight the scientific rationale and clinical experience and discuss ongoing trials using pancreatic cancer vaccines.


Oncologist | 2017

Comprehensive Genomic Profiling Aids in Distinguishing Metastatic Recurrence from Second Primary Cancers

Benjamin A. Weinberg; Kyle Gowen; Thomas K. Lee; Sai-Hong Ignatius Ou; Robert E. Bristow; Lauren S. Krill; M. Isabel Almira-Suarez; Siraj M. Ali; Vincent A. Miller; Stephen V. Liu; Samuel J. Klempner

BACKGROUND Metastatic recurrence after treatment for locoregional cancer is a major cause of morbidity and cancer-specific mortality. Distinguishing metastatic recurrence from the development of a second primary cancer has important prognostic and therapeutic value and represents a difficult clinical scenario. Advances beyond histopathological comparison are needed. We sought to interrogate the ability of comprehensive genomic profiling (CGP) to aid in distinguishing between these clinical scenarios. MATERIALS AND METHODS We identified three prospective cases of recurrent tumors in patients previously treated for localized cancers in which histologic analyses suggested subsequent development of a distinct second primary. Paired samples from the original primary and recurrent tumor were subjected to hybrid capture next-generation sequencing-based CGP to identify base pair substitutions, insertions, deletions, copy number alterations (CNA), and chromosomal rearrangements. Genomic profiles between paired samples were compared using previously established statistical clonality assessment software to gauge relatedness beyond global CGP similarities. RESULTS A high degree of similarity was observed among genomic profiles from morphologically distinct primary and recurrent tumors. Genomic information suggested reclassification as recurrent metastatic disease, and patients received therapy for metastatic disease based on the molecular determination. CONCLUSIONS Our cases demonstrate an important adjunct role for CGP technologies in separating metastatic recurrence from development of a second primary cancer. Larger series are needed to confirm our observations, but comparative CGP may be considered in patients for whom distinguishing metastatic recurrence from a second primary would alter the therapeutic approach. The Oncologist 2017;22:152-157Implications for Practice: Distinguishing a metastatic recurrence from a second primary cancer can represent a difficult clinicopathologic problem but has important prognostic and therapeutic implications. Approaches to aid histologic analysis may improve clinician and pathologist confidence in this increasingly common clinical scenario. Our series provides early support for incorporating paired comprehensive genomic profiling in clinical situations in which determination of metastatic recurrence versus a distinct second primary cancer would influence patient management.


Oncology | 2015

Current Standards and Novel Treatment Options for Metastatic Pancreatic Adenocarcinoma.

Benjamin A. Weinberg; Yabar Cs; Brody; Pishvaian Mj


Journal of Clinical Oncology | 2017

Characterization of tumor mutation burden (TMB) in gastrointestinal (GI) cancers.

Mohamed E. Salem; Joanne Xiu; Benjamin A. Weinberg; Wafik S. El-Deiry; Louis M. Weiner; Zoran Gatalica; Zhuqing Liu; Hesham El Ghazaly; Nianqing Xiao; Jimmy J. Hwang; Philip A. Philip; Anthony F. Shields; Heinz-Josef Lenz; John Marshall


Clinical advances in hematology & oncology | 2017

Evolving standards of care for resected pancreatic cancer.

Benjamin A. Weinberg; Philip Pa; Mohamed E. Salem


Clinical advances in hematology & oncology | 2016

A Paradigm Shift From One-Size-Fits-All to Tailor-Made Therapy for Metastatic Colorectal Cancer

Benjamin A. Weinberg; John L. Marshall; Marion L. Hartley; Mohamed E. Salem

Collaboration


Dive into the Benjamin A. Weinberg's collaboration.

Top Co-Authors

Avatar

Mohamed E. Salem

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

John L. Marshall

Georgetown University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jimmy J. Hwang

Carolinas Healthcare System

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael J. Pishvaian

Georgetown University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Joanne Xiu

Carolinas Healthcare System

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Heinz-Josef Lenz

University of Southern California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge