Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Cancer Res Commun ; 4(4): 1050-1062, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38592453

ABSTRACT

The ability to temporally regulate gene expression and track labeled cells makes animal models powerful biomedical tools. However, sudden expression of xenobiotic genes [e.g., GFP, luciferase (Luc), or rtTA3] can trigger inadvertent immunity that suppresses foreign protein expression or results in complete rejection of transplanted cells. Germline exposure to foreign antigens somewhat addresses these challenges; however, native fluorescence and bioluminescence abrogates the utility of reporter proteins and highly spatiotemporally restricted expression can lead to suboptimal xenoantigen tolerance. To overcome these unwanted immune responses and enable reliable cell tracking/gene regulation, we developed a novel mouse model that selectively expresses antigen-intact but nonfunctional forms of GFP and Luc, as well as rtTA3, after CRE-mediated recombination. Using tissue-specific CREs, we observed model and sex-based differences in immune tolerance to the encoded xenoantigens, illustrating the obstacles of tolerizing animals to foreign genes and validating the utility of these "NoGlow" mice to dissect mechanisms of central and peripheral tolerance. Critically, tissue unrestricted NoGlow mice possess no detectable background fluorescence or luminescence and exhibit limited adaptive immunity against encoded transgenic xenoantigens after vaccination. Moreover, we demonstrate that NoGlow mice allow tracking and tetracycline-inducible gene regulation of triple-transgenic cells expressing GFP/Luc/rtTA3, in contrast to transgene-negative immune-competent mice that eliminate these cells or prohibit metastatic seeding. Notably, this model enables de novo metastasis from orthotopically implanted, triple-transgenic tumor cells, despite high xenoantigen expression. Altogether, the NoGlow model provides a critical resource for in vivo studies across disciplines, including oncology, developmental biology, infectious disease, autoimmunity, and transplantation. SIGNIFICANCE: Multitolerant NoGlow mice enable tracking and gene manipulation of transplanted tumor cells without immune-mediated rejection, thus providing a platform to investigate novel mechanisms of adaptive immunity related to metastasis, immunotherapy, and tolerance.


Subject(s)
Antigens, Heterophile , Cell Tracking , Animals , Mice , Gene Expression Regulation , Mice, Transgenic , Disease Models, Animal
2.
Hum Vaccin Immunother ; 20(1): 2309693, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38330990

ABSTRACT

ER+ breast cancers (BC) are characterized by the elevated expression and signaling of estrogen receptor alpha (ESR1), which renders them sensitive to anti-endocrine therapy. While these therapies are clinically effective, prolonged treatment inevitably results in therapeutic resistance, which can occur through the emergence of gain-of-function mutations in ESR1. The central importance of ESR1 and development of mutated forms of ESR1 suggest that vaccines targeting these proteins could potentially be effective in preventing or treating endocrine resistance. To explore the potential of this approach, we developed several recombinant vaccines encoding different mutant forms of ESR1 (ESR1mut) and validated their ability to elicit ESR1-specific T cell responses. We then developed novel ESR1mut-expressing murine mammary cancer models to test the anti-tumor potential of ESR1mut vaccines. We found that these vaccines could suppress tumor growth, ESR1mut expression and estrogen signaling in vivo. To illustrate the applicability of these findings, we utilize HPLC to demonstrate the presentation of ESR1 and ESR1mut peptides on human ER+ BC cell MHC complexes. We then show the presence of human T cells reactive to ESR1mut epitopes in an ER+ BC patient. These findings support the development of ESR1mut vaccines, which we are testing in a Phase I clinical trial.


Subject(s)
Breast Neoplasms , Vaccines , Humans , Animals , Mice , Female , Breast Neoplasms/genetics , Breast Neoplasms/therapy , Mutation , Estrogens/therapeutic use , Signal Transduction , Vaccines/therapeutic use
3.
Hum Vaccin Immunother ; 19(3): 2291857, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-38087989

ABSTRACT

Carcinoembryonic antigen (CEA) is a glycosylated cell surface oncofetal protein involved in adhesion, proliferation, and migration that is highly upregulated in multiple carcinomas and has long been a promising target for cancer vaccination. This review summarizes the progress to date in the development of CEA vaccines, examining both pre-clinical and clinical studies across a variety of vaccine platforms that in aggregate, begin to reveal some critical insights. These studies demonstrate the ability of CEA vaccines to break immunologic tolerance and elicit CEA-specific immunity, which associates with improved clinical outcomes in select individuals. Approaches that have combined replicating viral vectors, with heterologous boosting and different adjuvant strategies have been particularly promising but, these early clinical trial results will require confirmatory studies. Collectively, these studies suggest that clinical efficacy likely depends upon harnessing a potent vaccine combination in an appropriate clinical setting to fully realize the potential of CEA vaccination.


Subject(s)
Cancer Vaccines , Neoplasms , Humans , Animals , Mice , Carcinoembryonic Antigen/genetics , Neoplasms/therapy , Genetic Vectors , Vaccination , Vaccines, Synthetic , Mice, Inbred C57BL
4.
JCI Insight ; 8(22)2023 Nov 22.
Article in English | MEDLINE | ID: mdl-37847565

ABSTRACT

Approximately 30% of breast cancer survivors deemed free of disease will experience locoregional or metastatic recurrence even up to 30 years after initial diagnosis, yet how residual/dormant tumor cells escape immunity elicited by the primary tumor remains unclear. We demonstrate that intrinsically dormant tumor cells are indeed recognized and lysed by antigen-specific T cells in vitro and elicit robust immune responses in vivo. However, despite close proximity to CD8+ killer T cells, dormant tumor cells themselves support early accumulation of protective FoxP3+ T regulatory cells (Tregs), which can be targeted to reduce tumor burden. These intrinsically dormant tumor cells maintain a hybrid epithelial/mesenchymal state that is associated with immune dysfunction, and we find that the tumor-derived, stem cell/basal cell protein Dickkopf WNT signaling pathway inhibitor 3 (DKK3) is critical for Treg inhibition of CD8+ T cells. We also demonstrate that DKK3 promotes immune-mediated progression of proliferative tumors and is significantly associated with poor survival and immunosuppression in human breast cancers. Together, these findings reveal that latent tumors can use fundamental mechanisms of tolerance to alter the T cell microenvironment and subvert immune detection. Thus, targeting these pathways, such as DKK3, may help render dormant tumors susceptible to immunotherapies.


Subject(s)
Breast Neoplasms , T-Lymphocytes, Regulatory , Humans , Female , T-Lymphocytes, Cytotoxic , Breast Neoplasms/pathology , Immunosuppression Therapy , Adaptive Immunity , Tumor Microenvironment , Adaptor Proteins, Signal Transducing/metabolism
5.
Clin Cancer Res ; 29(17): 3256-3258, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37428103

ABSTRACT

A vaccine targeting HER2, a nonmutated but overexpressed tumor antigen, readily primed T cells for ex vivo expansion and adoptive transfer with minimal toxicity. This regimen led to intramolecular epitope spreading in a majority of patients and offers a treatment modality that may improve outcomes for patients with metastatic breast cancer expressing HER2. See related article by Disis et al., p. 3362.


Subject(s)
Breast Neoplasms , Cancer Vaccines , Humans , Female , T-Lymphocytes/immunology , Breast Neoplasms/pathology , Cancer Vaccines/therapeutic use , Cancer Vaccines/immunology , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , Antigens, Neoplasm/immunology
6.
Cancer Gene Ther ; 30(6): 803-811, 2023 06.
Article in English | MEDLINE | ID: mdl-36765179

ABSTRACT

Therapeutic cancer vaccines, designed to activate immune effectors against tumor antigens, utilize a number of different platforms for antigen delivery. Among these are messenger RNAs (mRNA), successfully deployed in some prophylactic SARS-CoV2 vaccines. To enhance the immunogenicity of mRNA-delivered epitopes, self-replicating RNAs (srRNA) that markedly increase epitope expression have been developed. These vectors are derived from positive-strand RNA viruses in which the structural protein genes have been replaced with heterologous genes of interest, and the structural proteins are provided in trans to create single cycle viral replicon particles (VRPs). Clinical stage srRNA vectors have been derived from alphaviruses, including Venezuelan Equine Encephalitis (VEE), Sindbis, and Semliki Forest virus (SFV) and have encoded the tumor antigens carcinoembryonic antigen (CEA), human epidermal growth factor receptor 2 (HER2), prostate specific membrane antigen (PSMA), and human papilloma virus (HPV) antigens E6 and E7. Adverse events have mainly been grade 1 toxicities and minimal injection site reactions. We review here the clinical experience with these vaccines and our recent safety data from a study combining a VRP encoding HER2 plus an anti-PD1 monoclonal antibody (pembrolizumab). This experience with VRP-based srRNA supports recent development of fully synthetic srRNA technologies, where the viral structural proteins are replaced with protective lipid nanoparticles (LNP), cationic nanoemulsions or polymers.


Subject(s)
COVID-19 , Cancer Vaccines , Encephalitis Virus, Venezuelan Equine , Neoplasms , Humans , RNA, Viral/genetics , Cancer Vaccines/genetics , Encephalitis Virus, Venezuelan Equine/genetics , COVID-19/genetics , SARS-CoV-2/genetics , RNA, Messenger , Replicon , Genetic Vectors , Neoplasms/genetics , Neoplasms/therapy
7.
Cancer Gene Ther ; 30(6): 794-802, 2023 06.
Article in English | MEDLINE | ID: mdl-35821284

ABSTRACT

The development and success of RNA-based vaccines targeting SARS-CoV-2 has awakened new interest in utilizing RNA vaccines against cancer, particularly in the emerging use of self-replicating RNA (srRNA) viral vaccine platforms. These vaccines are based on different single-stranded RNA viruses, which encode RNA for target antigens in addition to replication genes that are capable of massively amplifying RNA messages after infection. The encoded replicase genes also stimulate innate immunity, making srRNA vectors ideal candidates for anti-tumor vaccination. In this review, we summarize different types of srRNA platforms that have emerged and review evidence for their efficacy in provoking anti-tumor immunity to different antigens. These srRNA platforms encompass the use of naked RNA, DNA-launched replicons, viral replicon particles (VRP), and most recently, synthetic srRNA replicon particles. Across these platforms, studies have demonstrated srRNA vaccine platforms to be potent inducers of anti-tumor immunity, which can be enhanced by homologous vaccine boosting and combining with chemotherapies, radiation, and immune checkpoint inhibition. As such, while this remains an active area of research, the past and present trajectory of srRNA vaccine development suggests immense potential for this platform in producing effective cancer vaccines.


Subject(s)
COVID-19 , Cancer Vaccines , Neoplasms , RNA Viruses , Humans , Genetic Vectors , Cancer Vaccines/genetics , Vaccination , SARS-CoV-2/genetics , RNA , RNA Viruses/genetics , Neoplasms/genetics , Neoplasms/therapy
8.
JCI Insight ; 7(6)2022 03 22.
Article in English | MEDLINE | ID: mdl-35167491

ABSTRACT

Two HER2-specific mAbs, trastuzumab and pertuzumab (T+P), combined with chemotherapy comprise standard-of-care treatment for advanced HER2+ breast cancers (BC). While this antibody combination is highly effective, its synergistic mechanism-of-action (MOA) remains incompletely understood. Past studies have suggested that the synergy underlying this combination occurs through the different mechanisms elicited by these antibodies, with pertuzumab suppressing HER2 heterodimerization and trastuzumab inducing antitumor immunity. However, in vivo evidence for this synergy is lacking. In this study, we found that the therapeutic efficacy elicited by their combination occurs through their joint ability to activate the classical complement pathway, resulting in both complement-dependent cytotoxicity and complement-dependent cellular phagocytosis of HER2+ tumors. We also demonstrate that tumor C1q expression is positively associated with survival outcome in HER2+ BC patients and that complement regulators CD55 and CD59 were inversely correlated with outcome, suggesting the clinical importance of complement activity. Accordingly, inhibition of C1q in mice abolished the synergistic therapeutic activity of T+P therapy, whereas knockdown of CD55 and CD59 expression enhanced T+P efficacy. In summary, our study identifies classical complement activation as a significant antitumor MOA for T+P therapy that may be functionally enhanced to potentially augment clinical therapeutic efficacy.


Subject(s)
Breast Neoplasms , Receptor, ErbB-2 , Animals , Antibodies, Monoclonal, Humanized , Breast Neoplasms/pathology , Cell Line, Tumor , Complement C1q , Female , Humans , Mice , Phagocytosis , Receptor, ErbB-2/metabolism , Trastuzumab/pharmacology , Trastuzumab/therapeutic use
9.
Mol Cancer Ther ; 21(1): 217-226, 2022 01.
Article in English | MEDLINE | ID: mdl-34675120

ABSTRACT

A noninvasive test to discriminate indolent prostate cancers from lethal ones would focus treatment where necessary while reducing overtreatment. We exploited the known activity of heat shock protein 90 (Hsp90) as a chaperone critical for the function of numerous oncogenic drivers, including the androgen receptor and its variants, to detect aggressive prostate cancer. We linked a near-infrared fluorescing molecule to an HSP90 binding drug and demonstrated that this probe (designated HS196) was highly sensitive and specific for detecting implanted prostate cancer cell lines with greater uptake by more aggressive subtypes. In a phase I human study, systemically administered HS196 could be detected in malignant nodules within prostatectomy specimens. Single-cell RNA sequencing identified uptake of HS196 by malignant prostate epithelium from the peripheral zone (AMACR+ERG+EPCAM+ cells), including SYP+ neuroendocrine cells that are associated with therapeutic resistance and metastatic progression. A theranostic version of this molecule is under clinical testing.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/genetics , Animals , Cell Line, Tumor , Humans , Male , Mice , Mice, SCID , Prostatic Neoplasms/pathology
11.
Cancer Res ; 81(18): 4641-4651, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34145037

ABSTRACT

Monoclonal antibodies (mAb) are a major component of cancer therapy. In this review, we summarize the different therapeutic mAbs that have been successfully developed against various tumor-expressed antigens and examine our current understanding of their different mechanisms of antitumor action. These mechanisms of action (MOA) largely center on the stimulation of different innate immune effector processes, which appear to be principally responsible for the efficacy of most unconjugated mAb therapies against cancer. This is evident in studies of mAbs targeting antigens for hematologic cancers, with emerging data also demonstrating the critical nature of innate immune-mediated mechanisms in the efficacy of anti-HER2 mAbs against solid HER2+ cancers. Although HER2-targeted mAbs were originally described as inhibitors of HER2-mediated signaling, multiple studies have since demonstrated these mAbs function largely through their engagement with Fc receptors to activate innate immune effector functions as well as complement activity. Next-generation mAbs are capitalizing on these MOAs through improvements to enhance Fc-activity, although regulation of these mechanisms may vary in different tumor microenvironments. In addition, novel antibody-drug conjugates have emerged as an important means to activate different MOAs. Although many unknowns remain, an improved understanding of these immunologic MOAs will be essential for the future of mAb therapy and cancer immunotherapy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Molecular Targeted Therapy , Neoplasms/drug therapy , Animals , Antibodies, Monoclonal/chemistry , Antineoplastic Agents, Immunological/chemistry , Biomarkers, Tumor , Clinical Decision-Making , Combined Modality Therapy , Disease Management , Disease Susceptibility , Hematologic Neoplasms/diagnosis , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/etiology , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Molecular Targeted Therapy/adverse effects , Molecular Targeted Therapy/methods , Neoplasms/etiology , Neoplasms/pathology , Prognosis , Treatment Outcome
12.
Mol Cancer Res ; 19(10): 1699-1711, 2021 10.
Article in English | MEDLINE | ID: mdl-34131071

ABSTRACT

HER2-positive breast cancers are among the most heterogeneous breast cancer subtypes. The early amplification of HER2 and its known oncogenic isoforms provide a plausible mechanism in which distinct programs of tumor heterogeneity could be traced to the initial oncogenic event. Here a Cancer rainbow mouse simultaneously expressing fluorescently barcoded wildtype (WTHER2), exon-16 null (d16HER2), and N-terminally truncated (p95HER2) HER2 isoforms is used to trace tumorigenesis from initiation to invasion. Tumorigenesis was visualized using whole-gland fluorescent lineage tracing and single-cell molecular pathology. We demonstrate that within weeks of expression, morphologic aberrations were already present and unique to each HER2 isoform. Although WTHER2 cells were abundant throughout the mammary ducts, detectable lesions were exceptionally rare. In contrast, d16HER2 and p95HER2 induced rapid tumor development. d16HER2 incited homogenous and proliferative luminal-like lesions which infrequently progressed to invasive phenotypes whereas p95HER2 lesions were heterogenous and invasive at the smallest detectable stage. Distinct cancer trajectories were observed for d16HER2 and p95HER2 tumors as evidenced by oncogene-dependent changes in epithelial specification and the tumor microenvironment. These data provide direct experimental evidence that intratumor heterogeneity programs begin very early and well in advance of screen or clinically detectable breast cancer. IMPLICATIONS: Although all HER2 breast cancers are treated equally, we show a mechanism by which clinically undetected HER2 isoforms program heterogenous cancer phenotypes through biased epithelial specification and adaptations within the tumor microenvironment.


Subject(s)
Breast Neoplasms/genetics , Carcinogenesis/genetics , Protein Isoforms/genetics , Receptor, ErbB-2/genetics , Animals , Female , Gene Expression Regulation, Neoplastic/genetics , Mice , Mice, Knockout , Tumor Microenvironment/genetics
13.
J Immunother Cancer ; 9(5)2021 05.
Article in English | MEDLINE | ID: mdl-33958486

ABSTRACT

BACKGROUND: Clinical studies have linked usage of progestins (synthetic progesterone [P4]) to breast cancer risk. However, little is understood regarding the role of native P4, signaling through the progesterone receptor (PR), in breast tumor formation. Recently, we reported a link between PR and immune signaling pathways, showing that P4/PR can repress type I interferon signaling pathways. Given these findings, we sought to investigate whether P4/PR drive immunomodulation in the mammary gland and promote tumor formation. METHODS: To determine the effect of P4 on immune cell populations in the murine mammary gland, mice were treated with P4 or placebo pellets for 21 days. Immune cell populations in the mammary gland, spleen, and inguinal lymph nodes were subsequently analyzed by flow cytometry. To assess the effect of PR overexpression on mammary gland tumor development as well as immune cell populations in the mammary gland, a transgenic mouse model was used in which PR was overexpressed throughout the entire mouse. Immune cell populations were assessed in the mammary glands, spleens, and inguinal lymph nodes of 6-month-old transgenic and control mice by flow cytometry. Transgenic mice were also monitored for mammary gland tumor development over a 2-year time span. Following development of mammary gland tumors, immune cell populations in the tumors and spleens of transgenic and control mice were analyzed by flow cytometry. RESULTS: We found that mice treated with P4 exhibited changes in the mammary gland indicative of an inhibited immune response compared with placebo-treated mice. Furthermore, transgenic mice with PR overexpression demonstrated decreased numbers of immune cell populations in their mammary glands, lymph nodes, and spleens. On long-term monitoring, we determined that multiparous PR-overexpressing mice developed significantly more mammary gland tumors than control mice. Additionally, tumors from PR-overexpressing mice contained fewer infiltrating immune cells. Finally, RNA sequencing analysis of tumor samples revealed that immune-related gene signatures were lower in tumors from PR-overexpressing mice as compared with control mice. CONCLUSION: Together, these findings offer a novel mechanism of P4-driven mammary gland tumor development and provide rationale in investigating the usage of antiprogestin therapies to promote immune-mediated elimination of mammary gland tumors.


Subject(s)
Breast Neoplasms/chemically induced , Cell Transformation, Neoplastic/chemically induced , Mammary Glands, Animal/drug effects , Progesterone/administration & dosage , Receptors, Progesterone/agonists , Tumor Escape/drug effects , Tumor Microenvironment/immunology , Adaptive Immunity/drug effects , Animals , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Drug Implants , Female , Galectin 4/genetics , Galectin 4/metabolism , Immunity, Innate/drug effects , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mammary Glands, Animal/immunology , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mice, Transgenic , Ovariectomy , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Signal Transduction , Time Factors , Tumor Burden/drug effects
15.
J Immunother Cancer ; 8(2)2020 11.
Article in English | MEDLINE | ID: mdl-33177177

ABSTRACT

BACKGROUND: There remains a significant need to eliminate the risk of recurrence of resected cancers. Cancer vaccines are well tolerated and activate tumor-specific immune effectors and lead to long-term survival in some patients. We hypothesized that vaccination with alphaviral replicon particles encoding tumor associated antigens would generate clinically significant antitumor immunity to enable prolonged overall survival (OS) in patients with both metastatic and resected cancer. METHODS: OS was monitored for patients with stage IV cancer treated in a phase I study of virus-like replicon particle (VRP)-carcinoembryonic antigen (CEA), an alphaviral replicon particle encoding a modified CEA. An expansion cohort of patients (n=12) with resected stage III colorectal cancer who had completed their standard postoperative adjuvant chemotherapy was administered VRP-CEA every 3 weeks for a total of 4 immunizations. OS and relapse-free survival (RFS) were determined, as well as preimmunization and postimmunization cellular and humoral immunity. RESULTS: Among the patients with stage IV cancer, median follow-up was 10.9 years and 5-year survival was 17%, (95% CI 6% to 33%). Among the patients with stage III cancer, the 5-year RFS was 75%, (95%CI 40% to 91%); no deaths were observed. At a median follow-up of 5.8 years (range: 3.9-7.0 years) all patients were still alive. All patients demonstrated CEA-specific humoral immunity. Patients with stage III cancer had an increase in CD8 +TEM (in 10/12) and decrease in FOXP3 +Tregs (in 10/12) following vaccination. Further, CEA-specific, IFNγ-producing CD8+granzyme B+TCM cells were increased. CONCLUSIONS: VRP-CEA induces antigen-specific effector T cells while decreasing Tregs, suggesting favorable immune modulation. Long-term survivors were identified in both cohorts, suggesting the OS may be prolonged.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Colonic Neoplasms/drug therapy , Immunologic Memory/physiology , T-Lymphocytes, Regulatory/immunology , Colonic Neoplasms/mortality , Female , Humans , Male , Neoplasm Staging , Survival Analysis
16.
Clin Cancer Res ; 26(17): 4670-4681, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32732224

ABSTRACT

PURPOSE: Despite promising advances in breast cancer immunotherapy, augmenting T-cell infiltration has remained a significant challenge. Although neither individual vaccines nor immune checkpoint blockade (ICB) have had broad success as monotherapies, we hypothesized that targeted vaccination against an oncogenic driver in combination with ICB could direct and enable antitumor immunity in advanced cancers. EXPERIMENTAL DESIGN: Our models of HER2+ breast cancer exhibit molecular signatures that are reflective of advanced human HER2+ breast cancer, with a small numbers of neoepitopes and elevated immunosuppressive markers. Using these, we vaccinated against the oncogenic HER2Δ16 isoform, a nondriver tumor-associated gene (GFP), and specific neoepitopes. We further tested the effect of vaccination or anti-PD-1, alone and in combination. RESULTS: We found that only vaccination targeting HER2Δ16, a driver of oncogenicity and HER2-therapeutic resistance, could elicit significant antitumor responses, while vaccines targeting a nondriver tumor-specific antigen or tumor neoepitopes did not. Vaccine-induced HER2-specific CD8+ T cells were essential for responses, which were more effective early in tumor development. Long-term tumor control of advanced cancers occurred only when HER2Δ16 vaccination was combined with αPD-1. Single-cell RNA sequencing of tumor-infiltrating T cells revealed that while vaccination expanded CD8 T cells, only the combination of vaccine with αPD-1 induced functional gene expression signatures in those CD8 T cells. Furthermore, we show that expanded clones are HER2-reactive, conclusively demonstrating the efficacy of this vaccination strategy in targeting HER2. CONCLUSIONS: Combining oncogenic driver targeted vaccines with selective ICB offers a rational paradigm for precision immunotherapy, which we are clinically evaluating in a phase II trial (NCT03632941).


Subject(s)
Breast Neoplasms/therapy , Cancer Vaccines/administration & dosage , Immune Checkpoint Inhibitors/administration & dosage , Mammary Neoplasms, Experimental/therapy , Receptor, ErbB-2/immunology , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/pathology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Female , Humans , Immunotherapy/methods , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Vaccines, Combined/administration & dosage
17.
Cancer Res ; 80(15): 3088-3100, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32366475

ABSTRACT

IL26 is a unique amphipathic member of the IL10 family of cytokines that participates in inflammatory signaling through a canonical receptor pathway. It also directly binds DNA to facilitate cellular transduction and intracellular inflammatory signaling. Although IL26 has almost no described role in cancer, our in vivo screen of inflammatory and cytokine pathway genes revealed IL26 to be one of the most significant inflammatory mediators of mammary engraftment and lung metastatic growth in triple-negative breast cancer (TNBC). Examination of human breast cancers demonstrated elevated IL26 transcripts in TNBC specimens, specifically in tumor cells as well as in Th17 CD4+ T cells within clinical TNBC specimens. IL26 did not have an autocrine effect on human TNBC cells, but rather its effect on engraftment and growth in vivo required neutrophils. IL26 enhanced mouse-derived DNA induction of inflammatory cytokines, which were collectively important for mammary and metastatic lung engraftment. To neutralize this effect, we developed a novel IL26 vaccine to stimulate antibody production and suppress IL26-enhanced engraftment in vivo, suggesting that targeting this inflammatory amplifier could be a unique means to control cancer-promoting inflammation in TNBC and other autoimmune diseases. Thus, we identified IL26 as a novel key modulator of TNBC metastasis and a potential therapeutic target in TNBC as well as other diseases reliant upon IL26-mediated inflammatory stimulation. SIGNIFICANCE: These findings identify IL26 as a unique, clinically relevant, inflammatory amplifier that enhances TNBC engraftment and dissemination in association with neutrophils, which has potential as a therapeutic target. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/15/3088/F1.large.jpg.


Subject(s)
Cell Adhesion , Interleukins/physiology , Neoplasm Transplantation , Neutrophils/physiology , Triple Negative Breast Neoplasms/pathology , Animals , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cells, Cultured , DNA, Neoplasm/drug effects , DNA, Neoplasm/metabolism , Disease Progression , Extracellular Traps/drug effects , Extracellular Traps/metabolism , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Inflammation Mediators/pharmacology , Inflammation Mediators/physiology , Interleukins/genetics , Interleukins/pharmacology , Mice , Mice, Inbred C57BL , Mice, SCID , Neoplasm Transplantation/immunology , Neoplasm Transplantation/pathology , Neutrophils/pathology , Triple Negative Breast Neoplasms/genetics , Xenograft Model Antitumor Assays
18.
JCI Insight ; 4(24)2019 12 19.
Article in English | MEDLINE | ID: mdl-31689243

ABSTRACT

The HER2-specific monoclonal antibody (mAb), trastuzumab, has been the mainstay of therapy for HER2+ breast cancer (BC) for approximately 20 years. However, its therapeutic mechanism of action (MOA) remains unclear, with antitumor responses to trastuzumab remaining heterogeneous and metastatic HER2+ BC remaining incurable. Consequently, understanding its MOA could enable rational strategies to enhance its efficacy. Using both murine and human versions of trastuzumab, we found its antitumor activity dependent on Fcγ receptor stimulation of tumor-associated macrophages (TAMs) and antibody-dependent cellular phagocytosis (ADCP), but not cellular cytotoxicity (ADCC). Trastuzumab also stimulated TAM activation and expansion, but did not require adaptive immunity, natural killer cells, and/or neutrophils. Moreover, inhibition of the innate immune ADCP checkpoint, CD47, significantly enhanced trastuzumab-mediated ADCP and TAM expansion and activation, resulting in the emergence of a unique hyperphagocytic macrophage population, improved antitumor responses, and prolonged survival. In addition, we found that tumor-associated CD47 expression was inversely associated with survival in HER2+ BC patients and that human HER2+ BC xenografts treated with trastuzumab plus CD47 inhibition underwent complete tumor regression. Collectively, our study identifies trastuzumab-mediated ADCP as an important antitumor MOA that may be clinically enabled by CD47 blockade to augment therapeutic efficacy.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/drug effects , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , CD47 Antigen/antagonists & inhibitors , Phagocytosis/drug effects , Trastuzumab/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast/pathology , Breast Neoplasms/immunology , Breast Neoplasms/mortality , Breast Neoplasms/pathology , CD47 Antigen/immunology , CD47 Antigen/metabolism , Cell Line, Tumor , Drug Synergism , Female , Humans , Immunity, Innate/drug effects , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Phagocytosis/immunology , Prognosis , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Trastuzumab/therapeutic use , Xenograft Model Antitumor Assays
19.
Cell ; 178(4): 1016-1028.e13, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31398327

ABSTRACT

T cell recognition of specific antigens mediates protection from pathogens and controls neoplasias, but can also cause autoimmunity. Our knowledge of T cell antigens and their implications for human health is limited by the technical limitations of T cell profiling technologies. Here, we present T-Scan, a high-throughput platform for identification of antigens productively recognized by T cells. T-Scan uses lentiviral delivery of antigen libraries into cells for endogenous processing and presentation on major histocompatibility complex (MHC) molecules. Target cells functionally recognized by T cells are isolated using a reporter for granzyme B activity, and the antigens mediating recognition are identified by next-generation sequencing. We show T-Scan correctly identifies cognate antigens of T cell receptors (TCRs) from viral and human genome-wide libraries. We apply T-Scan to discover new viral antigens, perform high-resolution mapping of TCR specificity, and characterize the reactivity of a tumor-derived TCR. T-Scan is a powerful approach for studying T cell responses.


Subject(s)
Antigens, Neoplasm/immunology , Epitopes, T-Lymphocyte/immunology , Genes, MHC Class I/immunology , HLA Antigens/immunology , Neoplasm Proteins/immunology , Receptors, Antigen, T-Cell/immunology , Antigen Presentation/immunology , Antigens, Neoplasm/genetics , Blood Donors , CD8-Positive T-Lymphocytes/metabolism , Female , Gene Knockout Techniques , Genes, MHC Class I/genetics , Granzymes/metabolism , HEK293 Cells , HLA Antigens/genetics , Humans , Neoplasm Proteins/genetics , Transduction, Genetic , Transfection
20.
Clin Cancer Res ; 25(9): 2725-2736, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30635338

ABSTRACT

PURPOSE: Immune-based therapy for metastatic breast cancer has had limited success, particularly in molecular subtypes with low somatic mutations rates. Strategies to augment T-cell infiltration of tumors include vaccines targeting established oncogenic drivers such as the genomic amplification of HER2. We constructed a vaccine based on a novel alphaviral vector encoding a portion of HER2 (VRP-HER2). PATIENTS AND METHODS: In preclinical studies, mice were immunized with VRP-HER2 before or after implantation of hHER2+ tumor cells and HER2-specific immune responses and antitumor function were evaluated. We tested VRP-HER2 in a phase I clinical trial where subjects with advanced HER2-overexpressing malignancies in cohort 1 received VRP-HER2 every 2 weeks for a total of 3 doses. In cohort 2, subjects received the same schedule concurrently with a HER2-targeted therapy. RESULTS: Vaccination in preclinical models with VRP-HER2 induced HER2-specific T cells and antibodies while inhibiting tumor growth. VRP-HER2 was well tolerated in patients and vaccination induced HER2-specific T cells and antibodies. Although a phase I study, there was 1 partial response and 2 patients with continued stable disease. Median OS was 50.2 months in cohort 1 (n = 4) and 32.7 months in cohort 2 (n = 18). Perforin expression by memory CD8 T cells post-vaccination significantly correlated with improved PFS. CONCLUSIONS: VRP-HER2 increased HER2-specific memory CD8 T cells and had antitumor effects in preclinical and clinical studies. The expansion of HER2-specific memory CD8 T cells in vaccinated patients was significantly correlated with increased PFS. Subsequent studies will seek to enhance T-cell activity by combining with anti-PD-1.


Subject(s)
Breast Neoplasms/therapy , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Immunity, Humoral/immunology , Immunologic Memory/immunology , Receptor, ErbB-2/immunology , Vaccines, Subunit/administration & dosage , Adult , Aged , Alphavirus/genetics , Animals , Apoptosis , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cancer Vaccines/immunology , Cell Proliferation , Dendritic Cells/immunology , Female , Follow-Up Studies , Genetic Vectors/administration & dosage , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Prognosis , Receptor, ErbB-2/metabolism , Survival Rate , Tumor Cells, Cultured , Vaccines, Subunit/immunology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...