Network


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

Hotspot


Dive into the research topics where Murali Janakiram is active.

Publication


Featured researches published by Murali Janakiram.


Trends in Pharmacological Sciences | 2015

New immunotherapies targeting the PD-1 pathway

Jordan M. Chinai; Murali Janakiram; Fuxiang Chen; Wantao Chen; Mark Kaplan; Xingxing Zang

Ligands from the B7 family bind to receptors of the CD28 family, which regulate early T cell activation in lymphoid organs and control inflammation and autoimmunity in peripheral tissues. Programmed death-1 (PD-1), a member of the CD28 family, is an inhibitory receptor on T cells and is responsible for their dysfunction in infectious diseases and cancers. The complex mechanisms controlling the expression and signaling of PD-1 and programmed death ligand 1 (PD-L1) are emerging. Recently completed and ongoing clinical trials that target these molecules have shown remarkable success by generating durable clinical responses in some cancer patients. In chronic viral infections, preclinical data reveal that targeting PD-1 and its ligands can improve T cell responses and virus clearance. There is also promise in stimulating this pathway for the treatment of autoimmune and inflammatory disorders.


Clinical Cancer Research | 2015

Expression, Clinical Significance, and Receptor Identification of the Newest B7 Family Member HHLA2 Protein

Murali Janakiram; Jordan M. Chinai; Susan Fineberg; Andras Fiser; Cristina Montagna; Ramadevi Medavarapu; Ekaterina Castano; Hyungjun Jeon; Kim C. Ohaegbulam; Ruihua Zhao; Aimin Zhao; Steven C. Almo; Joseph A. Sparano; Xingxing Zang

Purpose: HHLA2 (B7H7/B7-H5/B7y) is a newly identified B7 family member that regulates human T-cell functions. However, its protein expression in human organs and significance in human diseases are unknown. The objective of this study was to analyze HHLA2 protein expression in normal human tissues and cancers, as well as its prognostic significance, to explore mechanisms regulating HHLA2 expression, and to identify candidate HHLA2 receptors. Experimental Design: An immunohistochemistry protocol and a flow cytometry assay with newly generated monoclonal antibodies were developed to examine HHLA2 protein. HHLA2 gene copy-number variation was analyzed from cancer genomic data. The combination of bioinformatics analysis and immunologic approaches was established to explore HHLA2 receptors. Results: HHLA2 protein was detected in trophoblastic cells of the placenta and the epithelium of gut, kidney, gallbladder, and breast, but not in most other organs. In contrast, HHLA2 protein was widely expressed in human cancers from the breast, lung, thyroid, melanoma, pancreas, ovary, liver, bladder, colon, prostate, kidney, and esophagus. In a cohort of 50 patients with stage I–III triple-negative breast cancer, 56% of patients had aberrant expression of HHLA2 on their tumors, and high HHLA2 expression was significantly associated with regional lymph node metastasis and stage. The Cancer Genome Atlas revealed that HHLA2 copy-number gains were present in 29% of basal breast cancers, providing a potential mechanism for increased HHLA2 protein expression in breast cancer. Finally, Transmembrane and Immunoglobulin Domain Containing 2 (TMIGD2) was identified as one of the receptors for HHLA2. Conclusions: Wide expression of HHLA2 in human malignancies, together with its association with poor prognostic factors and its T-cell coinhibitory capability, suggests that the HHLA2 pathway represents a novel immunosuppressive mechanism within the tumor microenvironment and an attractive target for human cancer therapy. Clin Cancer Res; 21(10); 2359–66. ©2014 AACR. See related commentary by Xiao and Freeman, p. 2201


Cell Reports | 2014

Structure and cancer immunotherapy of the B7 family member B7x.

Hyungjun Jeon; Vladimir Vigdorovich; Sarah C. Garrett-Thomson; Murali Janakiram; Udupi A. Ramagopal; Yael M. Abadi; Jun Sik Lee; Lisa Scandiuzzi; Kim C. Ohaegbulam; Jordan M. Chinai; Ruihua Zhao; Yu Yao; Ying Mao; Joseph A. Sparano; Steven C. Almo; Xingxing Zang

B7x (B7-H4 or B7S1) is a member of the B7 family that can inhibit T cell function. B7x protein is absent in most normal human tissues and immune cells, but it is overexpressed in human cancers and often correlates with negative clinical outcome. The expression pattern and function of B7x suggest that it may be a potent immunosuppressive pathway in human cancers. Here, we determined the crystal structure of the human B7x immunoglobulin variable (IgV) domain at 1.59 Å resolution and mapped the epitopes recognized by monoclonal antibodies. We developed an in vivo system to screen therapeutic monoclonal antibodies against B7x and found that the clone 1H3 significantly inhibited growth of B7x-expressing tumors in vivo via multiple mechanisms. Furthermore, the surviving mice given 1H3 treatment were resistant to tumor rechallenge. Our data suggest that targeting B7x on tumors is a promising cancer immunotherapy and humanized 1H3 may be efficacious for immunotherapy of human cancers.


Immunotherapy | 2016

Immune checkpoint blockade in human cancer therapy: lung cancer and hematologic malignancies

Murali Janakiram; Vipul Pareek; Haiying Cheng; Deepa M. Narasimhulu; Xingxing Zang

Tumor immune evasion is one of the hallmarks of cancer, and expression of the B7 family of immune checkpoints (PD-L1, PD-L2, B7-H3, B7x and HHLA2) is one mechanism of immune evasion by tumors to suppress T-cell function. Antibodies blocking these interactions of B7-1/B7-2/CTLA-4 and PD-L1/PD-L2/PD-1 have had remarkable clinical success in several cancers and are less toxic than traditional chemotherapy. Even though only a small proportion of patients respond to checkpoint blockade, the duration of such responders due to immunological memory is remarkable and is longer than would be expected with any other agent in refractory disease. In this article, we review the therapeutic trials of blocking these pathways in human lung cancer and hematological malignancies.


Immunological Reviews | 2017

The third group of the B7‐CD28 immune checkpoint family: HHLA2, TMIGD2, B7x, and B7‐H3

Murali Janakiram; Urvi A. Shah; Weifeng Liu; Aimin Zhao; Mark P. Schoenberg; Xingxing Zang

The B7‐CD28 family of ligands and receptors play important roles in T‐cell co‐stimulation and co‐inhibition. Phylogenetically they can be divided into three groups. The recent discovery of the new molecules (B7‐H3 [CD276], B7x [B7‐H4/B7S1], and HHLA2 [B7H7/B7‐H5]/TMIGD2 [IGPR‐1/CD28H]) of the group III has expanded therapeutic possibilities for the treatment of human diseases. In this review, we describe the discovery, structure, and function of B7‐H3, B7x, HHLA2, and TMIGD2 in immune regulation. We also discuss their roles in important pathological states such as cancers, autoimmune diseases, transplantation, and infection. Various immunotherapeutical approaches are emerging including antagonistic monoclonal antibodies and agonistic fusion proteins to inhibit or potentiate these molecules and pathways in cancers and autoimmune diseases.


Clinical Cancer Research | 2017

HHLA2, a New Immune Checkpoint Member of the B7 Family, Is Widely Expressed in Human Lung Cancer and Associated with EGFR Mutational Status

Haiying Cheng; Murali Janakiram; Alain C. Borczuk; Juan Lin; Wanglong Qiu; Huijie Liu; Jordan M. Chinai; Balazs Halmos; Roman Perez-Soler; Xingxing Zang

Purpose: Immunotherapy with antibodies against B7/CD28 family members, including PD-1, PD-L1, and CTLA-4 has shifted the treatment paradigm for non–small cell lung carcinoma (NSCLC) with improved clinical outcome. HHLA2 is a newly discovered member of the family. By regulating T-cell function, HHLA2 could contribute to tumor immune suppression and thus be a novel target for cancer immunotherapy. There is limited information and critical need to characterize its expression profile and clinical significance in NSCLC. Experimental Design: We performed IHC with an HHLA2-specific antibody (clone 566.1) using tissue microarrays constructed from 679 NSCLC tumor tissues, including 392 cases in the discovery set and 287 cases in the validation cohort. We also studied clinicopathologic characteristics of these patients. Results: Overall, HHLA2 was not detected in most of normal lung tissue but expressed in 66% of NSCLC across different subtypes. In particular, EGFR-mutated NSCLC was significantly associated with higher tumor HHLA2 expression in both discovery (EGFR vs. WT: 76% vs. 53%, P = 0.01) and validation cohorts (89% vs. 69%, P = 0.01). In one of the two cohorts, HHLA2 expression was higher in lung adenocarcinoma as compared with squamous and large cell histology, non-Hispanic White versus Hispanics, and tumors with high tumor-infiltrating lymphocyte (TIL) density. In the multivariate analysis, EGFR mutation status and high TIL intensity were independently associated with HHLA2 expression in lung adenocarcinoma. Conclusions: HHLA2 is widely expressed in NSCLC and is associated with EGFR mutation and high TILs in lung adenocarcinoma. It is potentially a novel target for lung cancer immunotherapy. Clin Cancer Res; 23(3); 825–32. ©2016 AACR.


OncoImmunology | 2015

HHLA2 and TMIGD2: new immunotherapeutic targets of the B7 and CD28 families

Murali Janakiram; Jordan M. Chinai; Aimin Zhao; Joseph A. Sparano; Xingxing Zang

We and others recently discovered HHLA2 as a new B7 family member and transmembrane and immunoglobulin domain containing 2 (TMIGD2) as one of its receptors. Based on a new study we propose that HHLA2 may represent a novel immunosuppressive mechanism within the tumor microenvironment and hence could be a target for cancer therapy. TMIGD2 may be another therapeutic target.


The New England Journal of Medicine | 2018

Rapid progression of adult T-cell leukemia–lymphoma after PD-1 inhibitor therapy

Lee Ratner; Thomas A. Waldmann; Murali Janakiram; Jonathan E. Brammer

Progression of T-Cell Leukemia with Nivolumab Three patients with chronic, smoldering, and acute subtypes of adult T-cell leukemia–lymphoma had rapid disease progression and increased viral replica...


Thrombosis | 2013

A systematic review of the utility of residual vein obstruction studies in primary and secondary venous thrombosis.

Murali Janakiram; Matthew Sullivan; Marina Shcherba; Shuang Guo; Henny H. Billett

Background. Residual vein obstruction (RVO), the persistence of venous thrombosis with time and often after anticoagulation, may indicate a systemic prothrombotic condition. Prior studies have shown varying efficacy in using RVO as a risk factor for future venous thromboembolic (VTE) recurrence. Methods. To assess whether positive RVO imaging predicts recurrent VTE events, we performed a meta-analysis on studies in which patients with documented VTEs, anticoagulated for a minimum of 4 weeks, had repeat sonography to assess RVO and were subsequently followed for recurrent events. Results. Thirteen studies met inclusion criteria: 3531 patient VTE events with 3474 evaluable results were analyzed. The presence of RVO was associated with recurrence in all VTE (OR 1.93; 95% CI: 1.29, 2.89) and secondary VTE (OR 2.78; 95% CI: 1.41, 5.5) but not for primary VTE (OR 1.35; 95% CI: 0.87, 2.08). When cancer patients were eliminated from the secondary VTE group, there was no longer a significant association of RVO with VTE recurrence (OR 1.73; 95% CI: 0.81, 3.67) while in the subset of cancer patients, presence of RVO was associated with an increase in VTE recurrence risk (OR 5.14; 95% CI: 1.59, 16.65, P < 0.006). Conclusions. We conclude that the presence of RVO is associated with recurrence in secondary VTE but not in primary VTE and that association may be driven by the subset with cancer.


Leukemia | 2016

High prevalence and allele burden-independent prognostic importance of p53 mutations in an inner-city MDS/AML cohort

S Goel; J Hall; Kith Pradhan; Cassandra M. Hirsch; Bartlomiej Przychodzen; Aditi Shastri; Ioannis Mantzaris; Murali Janakiram; R Battini; N Kornblum; O Derman; K Gritsman; J Al-Hafidh; Y Wang; B Halmos; Ulrich Steidl; Jaroslaw P. Maciejewski; Ira Braunschweig; Amit Verma

High prevalence and allele burden-independent prognostic importance of p53 mutations in an inner-city MDS/AML cohort

Collaboration


Dive into the Murali Janakiram's collaboration.

Top Co-Authors

Avatar

Amit Verma

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Ira Braunschweig

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Aditi Shastri

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Ioannis Mantzaris

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Noah Kornblum

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Olga Derman

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Xingxing Zang

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Ramakrishna Battini

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Amer Assal

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Jordan M. Chinai

Albert Einstein College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge