Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 596(7870): 126-132, 2021 08.
Article in English | MEDLINE | ID: mdl-34290408

ABSTRACT

PD-1 blockade unleashes CD8 T cells1, including those specific for mutation-associated neoantigens (MANA), but factors in the tumour microenvironment can inhibit these T cell responses. Single-cell transcriptomics have revealed global T cell dysfunction programs in tumour-infiltrating lymphocytes (TIL). However, the majority of TIL do not recognize tumour antigens2, and little is known about transcriptional programs of MANA-specific TIL. Here, we identify MANA-specific T cell clones using the MANA functional expansion of specific T cells assay3 in neoadjuvant anti-PD-1-treated non-small cell lung cancers (NSCLC). We use their T cell receptors as a 'barcode' to track and analyse their transcriptional programs in the tumour microenvironment using coupled single-cell RNA sequencing and T cell receptor sequencing. We find both MANA- and virus-specific clones in TIL, regardless of response, and MANA-, influenza- and Epstein-Barr virus-specific TIL each have unique transcriptional programs. Despite exposure to cognate antigen, MANA-specific TIL express an incompletely activated cytolytic program. MANA-specific CD8 T cells have hallmark transcriptional programs of tissue-resident memory (TRM) cells, but low levels of interleukin-7 receptor (IL-7R) and are functionally less responsive to interleukin-7 (IL-7) compared with influenza-specific TRM cells. Compared with those from responding tumours, MANA-specific clones from non-responding tumours express T cell receptors with markedly lower ligand-dependent signalling, are largely confined to HOBIThigh TRM subsets, and coordinately upregulate checkpoints, killer inhibitory receptors and inhibitors of T cell activation. These findings provide important insights for overcoming resistance to PD-1 blockade.


Subject(s)
Antigens, Neoplasm/immunology , Carcinoma, Non-Small-Cell Lung/drug therapy , Gene Expression Regulation , Immune Checkpoint Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Antigens, Neoplasm/genetics , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Cells, Cultured , Humans , Immunologic Memory , Lung Neoplasms/genetics , Programmed Cell Death 1 Receptor/antagonists & inhibitors , RNA-Seq , Receptors, Interleukin-7/immunology , Single-Cell Analysis , Transcriptome/genetics , Tumor Microenvironment
2.
Clin Adv Hematol Oncol ; 22(5): 217-226, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38805313

ABSTRACT

Over the past decade, the lung cancer landscape has been dominated by targeted and immunotherapeutic approaches that have drastically shifted treatment paradigms for patients with advanced non-small cell lung cancer (NSCLC). Despite these scientific and clinical advances, there are still many unmet needs underscoring the importance of novel strategies. Antibody-drug conjugates (ADCs) represent one such strategy that is beginning to alter the therapeutic strategies for patients with advanced NSCLC. The rationale of ADCs is simple: selectively deliver cytotoxic payloads through an antibody-mediated process to target antigens expressed by cancer cells, sparing normal tissue and inflicting damage to tumors. Although this concept has been the leading view, preclinical and clinical observations are demonstrating that only a nascent mechanistic understanding of these agents exists. In this review, we discuss the underlying biology of ADCs and their structure and potential mechanisms of action, examine approved and promising ADC targets in lung cancer, and review emerging ADC targets and combinatorial strategies. Importantly, we address the unanswered questions surrounding ADCs in lung cancer, including biomarker selection, treatment sequencing, and mechanisms of resistance, as well as management of unique ADC-associated toxicities.


Subject(s)
Immunoconjugates , Lung Neoplasms , Humans , Immunoconjugates/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology
3.
Curr Oncol Rep ; 25(8): 913-922, 2023 08.
Article in English | MEDLINE | ID: mdl-37249833

ABSTRACT

PURPOSE OF REVIEW: For decades, early-stage resectable non-small cell lung cancer (NSCLC), while potentially curable, has been marred by unacceptably high recurrence rates. RECENT FINDINGS: Anti-PD(L)1 immune checkpoint blockade (ICB) has revolutionized the treatment of advanced NSCLC, and with recent approvals in the peri-operative space, is now poised to transform the systemic treatment paradigm for localized and locally-advanced NSCLC. In this review, we focus on neoadjuvant ICB in resectable NSCLC, highlighting the pre-clinical rationale for neoadjuvant ICB, early clinical trials, randomized phase 3 trial data, and future directions for resectable NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/surgery , Lung Neoplasms/drug therapy , Lung Neoplasms/surgery , Neoadjuvant Therapy , Immunotherapy , Clinical Trials, Phase III as Topic , Randomized Controlled Trials as Topic
4.
Cancer Immunol Immunother ; 71(10): 2405-2420, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35217892

ABSTRACT

Human gut microbial species found to associate with clinical responses to immune checkpoint inhibitors (ICIs) are often tested in mice using fecal microbiota transfer (FMT), wherein tumor responses in recipient mice may recapitulate human responses to ICI treatment. However, many FMT studies have reported only limited methodological description, details of murine cohorts, and statistical methods. To investigate the reproducibility and robustness of gut microbial species that impact ICI responses, we performed human to germ-free mouse FMT using fecal samples from patients with non-small cell lung cancer who had a pathological response or nonresponse after neoadjuvant ICI treatment. R-FMT mice yielded greater anti-tumor responses in combination with anti-PD-L1 treatment compared to NR-FMT, although the magnitude varied depending on mouse cell line, sex, and individual experiment. Detailed investigation of post-FMT mouse microbiota using 16S rRNA amplicon sequencing, with models to classify and correct for biological variables, revealed a shared presence of the most highly abundant taxa between the human inocula and mice, though low abundance human taxa colonized mice more variably after FMT. Multiple Clostridium species also correlated with tumor outcome in individual anti-PD-L1-treated R-FMT mice. RNAseq analysis revealed differential expression of T and NK cell-related pathways in responding tumors, irrespective of FMT source, with enrichment of these cell types confirmed by immunohistochemistry. This study identifies several human gut microbial species that may play a role in clinical responses to ICIs and suggests attention to biological variables is needed to improve reproducibility and limit variability across experimental murine cohorts.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Animals , Fecal Microbiota Transplantation , Humans , Mice , Neoadjuvant Therapy , RNA, Ribosomal, 16S/genetics , Reproducibility of Results
5.
Curr Oncol Rep ; 24(12): 1829-1841, 2022 12.
Article in English | MEDLINE | ID: mdl-36197593

ABSTRACT

PURPOSE OF REVIEW: Antibody-drug conjugates (ADCs) are a class of therapeutics that combine target-specific monoclonal antibodies with cytotoxic chemotherapy. Here, we describe the components of ADCs and review their promising activity, safety, and applicability in non-small cell lung cancer (NSCLC). RECENT FINDINGS: Technological advancements have reinvigorated ADCs as a viable treatment strategy in advanced solid tumors. Several target-specific ADCs have shown promise in treatment-refractory NSCLC, including agents targeting HER2, HER3, TROP2, CEACAM5, and MET, among others, with multiple confirmatory phase 3 trials ongoing. Critically, ADCs have demonstrated efficacy signals in both driver mutation-positive and mutation-negative advanced NSCLC, reinforcing their potential as an efficacious treatment strategy that transcends diverse tumor biology in advanced NSCLC. ADCs are a promising class of anti-cancer therapeutics that have significant potential in advanced NSCLC. Beyond confirmatory phase 3 trials, several questions remain including optimal agent sequencing, combinatorial methods, and unique toxicity management.


Subject(s)
Antineoplastic Agents, Immunological , Antineoplastic Agents , Carcinoma, Non-Small-Cell Lung , Immunoconjugates , Lung Neoplasms , Humans , Immunoconjugates/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Antineoplastic Agents/therapeutic use , Lung Neoplasms/drug therapy , Antineoplastic Agents, Immunological/therapeutic use
7.
Annu Rev Med ; 70: 425-435, 2019 01 27.
Article in English | MEDLINE | ID: mdl-30355264

ABSTRACT

Early-stage non-small cell lung cancer is a potentially curable disease, but with relapse rates exceeding 50% with standard treatments, this is a patient population in critical need of therapy innovation. Immunotherapy with immune checkpoint blockade has revolutionized the treatment strategy for advanced lung cancer. However, the role of this therapy in earlier-stage disease is largely unknown. The study of immunotherapy in earlier-stage disease has many advantages, including assessment of pathologic response and incorporation of translational scientific analyses to evaluate antitumor immune responses. Multiple clinical trials are currently under way, with promising early results.


Subject(s)
Antineoplastic Agents, Immunological/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Nivolumab/administration & dosage , Programmed Cell Death 1 Receptor/drug effects , Animals , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/surgery , Combined Modality Therapy , Disease-Free Survival , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Lung Neoplasms/surgery , Neoadjuvant Therapy , Neoplasm Invasiveness/pathology , Neoplasm Staging , Pneumonectomy/methods , Prognosis , Risk Assessment , Survival Rate , Treatment Outcome
8.
Curr Oncol Rep ; 22(6): 56, 2020 05 16.
Article in English | MEDLINE | ID: mdl-32415399

ABSTRACT

PURPOSE OF REVIEW: Checkpoint inhibitor pneumonitis (CIP) is a toxicity of immune checkpoint blockade (ICB) that can be highly morbid and at times fatal. Here, we review the proposed biologic mechanisms of CIP, epidemiology and risk factors for CIP development, diagnostic work-up and management strategies for CIP, and future directions of CIP research. RECENT FINDINGS: CIP incidence appears to be greater in real-world populations and may continue to rise as FDA approvals for ICB continue to expand to multiple malignancies. Multiple retrospective studies and case series have identified potential risk factors for CIP. Several society guidelines have helped to unify the classification of CIP severity and standardize treatment approaches but significant gaps remain, including formal validated diagnostic criteria for CIP. While significant strides have been made in enhancing the knowledge and management of CIP, ongoing research is needed to continue to advance our understanding of the biologic underpinnings of CIP, as well as optimize diagnostic and management strategies for this potentially devastating toxicity.


Subject(s)
Immune Checkpoint Inhibitors/adverse effects , Pneumonia/chemically induced , Humans , Pneumonia/diagnosis , Pneumonia/therapy , Retrospective Studies
9.
Clin Adv Hematol Oncol ; 18(9): 562-572, 2020 Sep.
Article in English | MEDLINE | ID: mdl-33006585

ABSTRACT

Therapeutic advancement for mesothelioma has been stagnant, with minimal treatment innovation in the past decade. Recently, however, immune checkpoint blockade (ICB) targeting the programmed death 1 and cytotoxic T-lymphocyte-associated antigen 4 pathways has revolutionized the treatment of multiple malignancies and shown promise in mesothelioma, with multiple agents now recommended in the salvage setting for advanced disease progressive on platinum-based chemotherapy. Studies of frontline chemoimmunotherapy and ICB combinations have also been encouraging, and both are likely to become integrated into the frontline treatment strategy for mesothelioma in the coming years. Other novel immunotherapy strategies, including chimeric antigen receptor T-cell therapy, are being investigated in mesothelioma. Although early studies have demonstrated the safety of multiple agents, further trials powered for efficacy are needed. In addition, enrolling patients in window-of-opportunity trials of ICB in resectable mesothelioma and biomarker-focused correlative studies will be critical to furthering the mechanistic understanding of ICB in mesothelioma, which in turn will help to uncover biomarkers of response and resistance in these patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Immunotherapy , Mesothelioma , Platinum/therapeutic use , CTLA-4 Antigen/immunology , Combined Modality Therapy , Humans , Mesothelioma/immunology , Mesothelioma/therapy , Neoplasm Proteins , Platinum/immunology , Programmed Cell Death 1 Receptor/immunology
10.
Anticancer Drugs ; 30(9): 879-885, 2019 10.
Article in English | MEDLINE | ID: mdl-31503012

ABSTRACT

Inflammation is essential for our innate and adaptive immunity, but chronic inflammation can also be detrimental, playing a role in tumor development and subversion of host immunity. A multitude of proteins and cytokines are involved in chronic inflammation; interleukin-1ß, in particular, has been recognized as a critical pro-inflammatory cytokine that can trigger a cascade of inflammatory mediators, promoting angiogenesis, tumor invasiveness, and metastasis. The inhibition of interleukin-1ß with the antibody canakinumab was recently highlighted in a large-scale trial studying the effects of the inflammatory modulating antibody in heart disease. In this study, a marked decrease in the incidence of lung cancer (a 67% relative risk reduction) was observed in a high-risk population. Although a number of preclinical studies have demonstrated that canakinumab inhibits interleukin-1ß and reduces inflammation, the question remains whether these actions positively affect both cancer incidence and recurrence. This review will summarize the role of inflammation in cancer propagation and development, discuss the biological rationale for targeting interleukin-1ß in lung cancer, advocate for further investigation of the anti-inflammatory antibody canakinumab as a new attractive mechanism for future lung cancer therapy, and discuss future and ongoing trials.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Animals , Carcinoma, Non-Small-Cell Lung/metabolism , Humans , Incidence , Inflammation/metabolism , Interleukin-1beta/metabolism , Lung Neoplasms/metabolism , Neoplasm Recurrence, Local/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL