Try a new search

Format these results:

Searched for:

person:bhardn02

Total Results:

252


Structural basis for self-discrimination by neoantigen-specific TCRs

Finnigan, John P; Newman, Jenna H; Patskovsky, Yury; Patskovska, Larysa; Ishizuka, Andrew S; Lynn, Geoffrey M; Seder, Robert A; Krogsgaard, Michelle; Bhardwaj, Nina
T cell receptors (TCR) are pivotal in mediating tumour cell cytolysis via recognition of mutation-derived tumour neoantigens (neoAgs) presented by major histocompatibility class-I (MHC-I). Understanding the factors governing the emergence of neoAg from somatic mutations is a major focus of current research. However, the structural and cellular determinants controlling TCR recognition of neoAgs remain poorly understood. This study describes the multi-level analysis of a model neoAg from the B16F10 murine melanoma, H2-Db/Hsf2 p.K72N68-76, as well as its cognate TCR 47BE7. Through cellular, molecular and structural studies we demonstrate that the p.K72N mutation enhances H2-Db binding, thereby improving cell surface presentation and stabilizing the TCR 47BE7 epitope. Furthermore, TCR 47BE7 exhibited high functional avidity and selectivity, attributable to a broad, stringent, binding interface enabling recognition of native B16F10 despite low antigen density. Our findings provide insight into the generation of anchor-residue modified neoAg, and emphasize the value of molecular and structural investigations of neoAg in diverse MHC-I contexts for advancing the understanding of neoAg immunogenicity.
PMID: 38459027
ISSN: 2041-1723
CID: 5645662

Lung Cancer and Severe Acute Respiratory Syndrome Coronavirus 2 Infection: Identifying Important Knowledge Gaps for Investigation

Rolfo, Christian; Meshulami, Noy; Russo, Alessandro; Krammer, Florian; García-Sastre, Adolfo; Mack, Philip C; Gomez, Jorge E; Bhardwaj, Nina; Benyounes, Amin; Sirera, Rafael; Moore, Amy; Rohs, Nicholas; Henschke, Claudia I; Yankelevitz, David; King, Jennifer; Shyr, Yu; Bunn, Paul A; Minna, John D; Hirsch, Fred R
Patients with lung cancer are especially vulnerable to coronavirus disease 2019 (COVID-19) with a greater than sevenfold higher rate of becoming infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) COVID-19, a greater than threefold higher hospitalization rate with high complication rates, and an estimated case fatality rate of more than 30%. The reasons for the increased vulnerability are not known. In addition, beyond the direct impact of the pandemic on morbidity and mortality among patients with lung cancer, COVID-19, with its disruption of patient care, has also resulted in substantial impact on lung cancer screening and treatment/management.COVID-19 vaccines are safe and effective in people with lung cancer. On the basis of the available data, patients with lung cancer should continue their course of cancer treatment and get vaccinated against the SARS-CoV-2 virus. For unknown reasons, some patients with lung cancer mount poor antibody responses to vaccination. Thus, boosting vaccination seems urgently indicated in this subgroup of vulnerable patients with lung cancer. Nevertheless, many unanswered questions regarding vaccination in this population remain, including the magnitude, quality, and duration of antibody response and the role of innate and acquired cellular immunities for clinical protection. Additional important knowledge gaps also remain, including the following: how can we best protect patients with lung cancer from developing COVID-19, including managing care in patient with lung cancer and the home environment of patients with lung cancer; are there clinical/treatment demographics and tumor molecular demographics that affect severity of COVID-19 disease in patients with lung cancer; does anticancer treatment affect antibody production and protection; does SARS-CoV-2 infection affect the development/progression of lung cancer; and are special measures and vaccine strategies needed for patients with lung cancer as viral variants of concern emerge.
PMCID:8579698
PMID: 34774792
ISSN: 1556-1380
CID: 5079242

Therapeutic cancer vaccines

Saxena, Mansi; van der Burg, Sjoerd H; Melief, Cornelis J M; Bhardwaj, Nina
Therapeutic cancer vaccines have undergone a resurgence in the past decade. A better understanding of the breadth of tumour-associated antigens, the native immune response and development of novel technologies for antigen delivery has facilitated improved vaccine design. The goal of therapeutic cancer vaccines is to induce tumour regression, eradicate minimal residual disease, establish lasting antitumour memory and avoid non-specific or adverse reactions. However, tumour-induced immunosuppression and immunoresistance pose significant challenges to achieving this goal. In this Review, we deliberate on how to improve and expand the antigen repertoire for vaccines, consider developments in vaccine platforms and explore antigen-agnostic in situ vaccines. Furthermore, we summarize the reasons for failure of cancer vaccines in the past and provide an overview of various mechanisms of resistance posed by the tumour. Finally, we propose strategies for combining suitable vaccine platforms with novel immunomodulatory approaches and standard-of-care treatments for overcoming tumour resistance and enhancing clinical efficacy.
PMID: 33907315
ISSN: 1474-1768
CID: 4895182

CSF1R inhibition depletes tumor-associated macrophages and attenuates tumor progression in a mouse sonic Hedgehog-Medulloblastoma model

Tan, I-Li; Arifa, Raquel Duque Nascimento; Rallapalli, Harikrishna; Kana, Veronika; Lao, Zhimin; Sanghrajka, Reeti Mayur; Sumru Bayin, N; Tanne, Antoine; Wojcinski, Alexandre; Korshunov, Andrey; Bhardwaj, Nina; Merad, Miriam; Turnbull, Daniel H; Lafaille, Juan J; Joyner, Alexandra L
The immune microenvironment of tumors can play a critical role in promoting or inhibiting tumor progression depending on the context. We present evidence that tumor-associated macrophages/microglia (TAMs) can promote tumor progression in the sonic hedgehog subgroup of medulloblastoma (SHH-MB). By combining longitudinal manganese-enhanced magnetic resonance imaging (MEMRI) and immune profiling of a sporadic mouse model of SHH-MB, we found the density of TAMs is higher in the ~50% of tumors that progress to lethal disease. Furthermore, reducing regulatory T cells or eliminating B and T cells in Rag1 mutants does not alter SHH-MB tumor progression. As TAMs are a dominant immune component in tumors and are normally dependent on colony-stimulating factor 1 receptor (CSF1R), we treated mice with a CSF1R inhibitor, PLX5622. Significantly, PLX5622 reduces a subset of TAMs, prolongs mouse survival, and reduces the volume of most tumors within 4 weeks of treatment. Moreover, concomitant with a reduction in TAMs the percentage of infiltrating cytotoxic T cells is increased, indicating a change in the tumor environment. Our studies in an immunocompetent preclinical mouse model demonstrate TAMs can have a functional role in promoting SHH-MB progression. Thus, CSF1R inhibition could have therapeutic potential for a subset of SHH-MB patients.
PMID: 33159168
ISSN: 1476-5594
CID: 4664582

Shared Immunogenic Poly-Epitope Frameshift Mutations in Microsatellite Unstable Tumors

Roudko, Vladimir; Bozkus, Cansu Cimen; Orfanelli, Theofano; McClain, Christopher B; Carr, Caitlin; O'Donnell, Timothy; Chakraborty, Lauren; Samstein, Robert; Huang, Kuan-Lin; Blank, Stephanie V; Greenbaum, Benjamin; Bhardwaj, Nina
Microsatellite instability-high (MSI-H) tumors are characterized by high tumor mutation burden and responsiveness to checkpoint blockade. We identified tumor-specific frameshifts encoding multiple epitopes that originated from indel mutations shared among patients with MSI-H endometrial, colorectal, and stomach cancers. Epitopes derived from these shared frameshifts have high population occurrence rates, wide presence in many tumor subclones, and are predicted to bind to the most frequent MHC alleles in MSI-H patient cohorts. Neoantigens arising from these mutations are distinctly unlike self and viral antigens, signifying novel groups of potentially highly immunogenic tumor antigens. We further confirmed the immunogenicity of frameshift peptides in T cell stimulation experiments using blood mononuclear cells isolated from both healthy donors and MSI-H cancer patients. Our study uncovers the widespread occurrence and strong immunogenicity of tumor-specific antigens derived from shared frameshift mutations in MSI-H cancer and Lynch syndrome patients, suitable for the design of common "off-the-shelf" cancer vaccines.
PMID: 33259803
ISSN: 1097-4172
CID: 4694092

Flt3 ligand augments immune responses to anti-DEC-205-NY-ESO-1 vaccine through expansion of dendritic cell subsets

Bhardwaj, Nina; Friedlander, Philip A; Pavlick, Anna C; Ernstoff, Marc S; Gastman, Brian R; Hanks, Brent A; Curti, Brendan D; Albertini, Mark R; Luke, Jason J; Blazquez, Ana B; Balan, Sreekumar; Bedognetti, Davide; Beechem, Joseph M; Crocker, Andrea S; D'Amico, Leonard; Danaher, Patrick; Davis, Thomas A; Hawthorne, Thomas; Hess, Bruce W; Keler, Tibor; Lundgren, Lisa; Morishima, Chihiro; Ramchurren, Nirasha; Rinchai, Darawan; Salazar, Andres M; Salim, Bob A; Sharon, Elad; Vitale, Laura A; Wang, Ena; Warren, Sarah; Yellin, Michael J; Disis, Mary L; Cheever, Martin A; Fling, Steven P
Generating responses to tumor antigens poses a challenge for immunotherapy. This phase II trial (NCT02129075) tested fms-like tyrosine kinase 3 (Flt3) ligand pre-treatment enhancement of responses to dendritic cell (DC)-targeting vaccines. We evaluated a regimen of Flt3L (CDX-301) to increase DCs and other antigen-presenting cells, poly-ICLC (TLR3 agonist that activates DCs) and a vaccine comprising anti-DEC-205-NY-ESO-1, a fusion antibody targeting CD205, linked to NY-ESO-1. High-risk melanoma patients were randomized to vaccine, with and without CDX-301. The end point was immune response to NY-ESO-1. Flt3L increased peripheral monocytes and conventional DCs (cDCs), including cross-presenting cDC1 and cDC2 and plasmacytoid DCs. Significant increases in humoral and T-cell responses and activation of DCs, natural killer cells and T cells were elicited. Transcriptional analyses revealed gene signatures associated with CDX-301 induction of an early, durable immune response. This study reveals in vivo effects of Flt3L on innate immune cells in the setting of vaccination, leading to an immunogenic vaccine regimen.
PMID: 35121932
ISSN: 2662-1347
CID: 5152862

Author Correction: A conserved dendritic-cell regulatory program limits antitumour immunity

Maier, Barbara; Leader, Andrew M; Chen, Steven T; Tung, Navpreet; Chang, Christie; LeBerichel, Jessica; Chudnovskiy, Aleksey; Maskey, Shrisha; Walker, Laura; Finnigan, John P; Kirkling, Margaret E; Reizis, Boris; Ghosh, Sourav; D'Amore, Natalie Roy; Bhardwaj, Nina; Rothlin, Carla V; Wolf, Andrea; Flores, Raja; Marron, Thomas; Rahman, Adeeb H; Kenigsberg, Ephraim; Brown, Brian D; Merad, Miriam
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
PMID: 32499658
ISSN: 1476-4687
CID: 4469402

A conserved dendritic-cell regulatory program limits antitumour immunity

Maier, Barbara; Leader, Andrew M; Chen, Steven T; Tung, Navpreet; Chang, Christie; LeBerichel, Jessica; Chudnovskiy, Aleksey; Maskey, Shrisha; Walker, Laura; Finnigan, John P; Kirkling, Margaret E; Reizis, Boris; Ghosh, Sourav; D'Amore, Natalie Roy; Bhardwaj, Nina; Rothlin, Carla V; Wolf, Andrea; Flores, Raja; Marron, Thomas; Rahman, Adeeb H; Kenigsberg, Ephraim; Brown, Brian D; Merad, Miriam
Checkpoint blockade therapies have improved cancer treatment, but such immunotherapy regimens fail in a large subset of patients. Conventional type 1 dendritic cells (DC1s) control the response to checkpoint blockade in preclinical models and are associated with better overall survival in patients with cancer, reflecting the specialized ability of these cells to prime the responses of CD8+ T cells1-3. Paradoxically, however, DC1s can be found in tumours that resist checkpoint blockade, suggesting that the functions of these cells may be altered in some lesions. Here, using single-cell RNA sequencing in human and mouse non-small-cell lung cancers, we identify a cluster of dendritic cells (DCs) that we name 'mature DCs enriched in immunoregulatory molecules' (mregDCs), owing to their coexpression of immunoregulatory genes (Cd274, Pdcd1lg2 and Cd200) and maturation genes (Cd40, Ccr7 and Il12b). We find that the mregDC program is expressed by canonical DC1s and DC2s upon uptake of tumour antigens. We further find that upregulation of the programmed death ligand 1 protein-a key checkpoint molecule-in mregDCs is induced by the receptor tyrosine kinase AXL, while upregulation of interleukin (IL)-12 depends strictly on interferon-γ and is controlled negatively by IL-4 signalling. Blocking IL-4 enhances IL-12 production by tumour-antigen-bearing mregDC1s, expands the pool of tumour-infiltrating effector T cells and reduces tumour burden. We have therefore uncovered a regulatory module associated with tumour-antigen uptake that reduces DC1 functionality in human and mouse cancers.
PMID: 32269339
ISSN: 1476-4687
CID: 4374592

Combined vaccination with NY-ESO-1 protein, poly-ICLC, and montanide improves humoral and cellular immune responses in high-risk melanoma patients

Pavlick, Anna; Blazquez, Ana B; Meseck, Marcia; Lattanzi, Michael; Ott, Patrick A; Marron, Thomas U; Holman, Rose Marie; Mandeli, John; Salazar, Andres M; McClain, Christopher B; Gimenez, Gustavo; Balan, Sreekumar; Gnjatic, Sacha; Sabado, Rachel Lubong; Bhardwaj, Nina
Given its ability to induce both humoral and cellular immune responses, NY-ESO-1 has been considered a suitable antigen for a cancer vaccine. Despite promising results from early-phase clinical studies in melanoma patients, NY-ESO-1 vaccine immunotherapy has not been widely investigated in larger trials; consequently, many questions remain as to the optimal vaccine formulation, predictive biomarkers, and sequencing and timing of vaccines in melanoma treatment. We conducted an adjuvant phase I/II clinical trial in high-risk resected melanoma to optimize the delivery of poly-ICLC, a TLR-3/MDA-5 agonist as a component of vaccine formulation. A phase I dose escalation part was undertaken to identify the maximum tolerated dose of poly-ICLC administered in combination with NY-ESO-1 and montanide. This was followed by a randomized phase II part investigating the maximum tolerated dose of poly-ICLC with NY-ESO-1 with or without montanide. The vaccine regimens were generally well-tolerated, with no treatment-related grade 3/4 adverse events. Both regimens induced integrated NY-ESO-1-specific CD4+ T-cell and humoral responses. CD8+ T-cell responses were mainly detected in patients receiving montanide. T-cell avidity towards NY-ESO-1 peptides was higher in patients vaccinated with montanide. In conclusion, NY-ESO-1 protein in combination with poly-ICLC is safe, well-tolerated, and capable of inducing integrated antibody and CD4+ T-cell responses in most patients. Combination with montanide enhances antigen specific T-cell avidity and CD8+ T-cell cross-priming in a fraction of patients, indicating that montanide contributes to the induction of specific CD8+ T-cell responses to NY-ESO-1.
PMID: 31699709
ISSN: 2326-6074
CID: 4179512

Poly-ICLC, a TLR3 Agonist, Induces Transient Innate Immune Responses in Patients With Treated HIV-Infection: A Randomized Double-Blinded Placebo Controlled Trial

Saxena, Mansi; Sabado, Rachel L; La Mar, Melissa; Mohri, Hiroshi; Salazar, Andres M; Dong, Hanqing; Correa Da Rosa, Joel; Markowitz, Martin; Bhardwaj, Nina; Miller, Elizabeth
Objective: Toll-like receptor-3 agonist Poly-ICLC has been known to activate immune cells and induce HIV replication in pre-clinical experiments. In this study we investigated if Poly-ICLC could be used for disrupting HIV latency while simultaneously enhancing innate immune responses. Design: This was a randomized, placebo-controlled, double-blinded trial in aviremic, cART-treated HIV-infected subjects. Participants (n = 15) were randomized 3:1 to receive two consecutive daily doses of Poly-ICLC (1.4 mg subcutaneously) vs. placebo. Subjects were observed for adverse events, immune activation, and viral replication. Methods: Besides primary outcomes of safety and tolerability, several longitudinal immune parameters were evaluated including immune cell phenotype and function via flowcytometry, ELISA, and transcriptional profiling. PCR assays for plasma HIV-1 RNA, CD4+ T cell-associated HIV-1 RNA, and proviral DNA were performed to measure HIV reservoirs and latency. Results: Poly-ICLC was overall safe and well-tolerated. Poly-ICLC-related adverse events were Grade 1/2, with the exception of one Grade 3 neutropenia which was short-lived. Mild Injection site reactions were observed in nearly all participants in the Poly-ICLC arm. Transcriptional analyses revealed upregulation of innate immune pathways in PBMCs following Poly-ICLC treatment, including strong interferon signaling accompanied by transient increases in circulating IP-10 (CXCL10) levels. These responses generally peaked by 24-48 h after the first injection and returned to baseline by day 8. CD4+ T cell number and phenotype were unchanged, plasma viral control was maintained and no significant effect on HIV reservoirs was observed. Conclusions: These finding suggest that Poly-ICLC could be safely used for inducing transient innate immune responses in treated HIV+ subjects indicating promise as an adjuvant for HIV therapeutic vaccines. Trial Registration: www.ClinicalTrials.gov, identifier: NCT02071095.
PMCID:6467168
PMID: 31024557
ISSN: 1664-3224
CID: 4453852