Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
2.
Clin Transl Oncol ; 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39046682

ABSTRACT

PURPOSE: Immunotherapy using immune checkpoint inhibitors (ICIs) has shown several benefits over traditional therapies. However, the eligible population remains small. Antibiotic (ATB) use might reduce immunotherapy efficacy by disrupting the gut microbiota. However, in China, ATB effect on ICI therapy efficacy remains unelucidated. We aimed to assess the effects of ATBs on the anti-tumor efficacy of ICIs to provide a reference for clinical use. METHODS: We included 134 patients with advanced tumors undergoing ICI therapy at Shanghai Jiading District Central Hospital from January 1, 2021, to October 1, 2023. They were divided into Non-ATB and ATB groups based on ATB use within 30 days before and after ICI administration. Moreover, we compared progression-free (PFS) and overall (OS) survival between the groups. RESULTS: Median PFS and OS were lower in the ATB than in the Non-ATB group (PFS: 4.0 vs. 5.5 months; OS: 5.4 vs. 6.5 months). Univariate analysis revealed that ATB use significantly affected PFS (hazard ratio [HR] = 2.318, 95% confidence interval [CI] = 1.281-4.194, P = 0.005) and OS (HR = 2.115, 95% CI = 1.161-3.850, P = 0.014). Moreover, multivariate analysis revealed poor PFS (HR = 2.573, 95% CI = 1.373-4.826, P = 0.003) and OS (HR = 2.452, 95% CI = 1.298-4.632, P = 0.006) in patients who received ATBs during ICI therapy. CONCLUSIONS: ATB use is negatively correlated with ICI therapy efficacy, leading to reduced PFS and OS in patients undergoing such treatment. Owing to the significant impact of ATBs on the human gut microbiome, regulation of the gut microbiome may emerge as a novel therapeutic target that can enhance the clinical activity of ICIs.

4.
Clin Transl Oncol ; 26(7): 1584-1612, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38512448

ABSTRACT

Glioblastoma multiform (GBM) is the most prevalent CNS (central nervous system) tumor in adults, with an average survival length shorter than 2 years and rare metastasis to organs other than CNS. Despite extensive attempts at surgical resecting, the inherently permeable nature of this disease has rendered relapse nearly unavoidable. Thus, immunotherapy is a feasible alternative, as stimulated immune cells can enter into the remote and inaccessible tumor cells. Immunotherapy has revolutionized patient upshots in various malignancies and might introduce different effective ways for GBM patients. Currently, researchers are exploring various immunotherapeutic strategies in patients with GBM to target both the innate and acquired immune responses. These approaches include reprogrammed tumor-associated macrophages, the use of specific antibodies to inhibit tumor progression and metastasis, modifying tumor-associated macrophages with antibodies, vaccines that utilize tumor-specific dendritic cells to activate anti-tumor T cells, immune checkpoint inhibitors, and enhanced T cells that function against tumor cells. Despite these findings, there is still room for improving the response faults of the many currently tested immunotherapies. This study aims to review the currently used immunotherapy approaches with their molecular mechanisms and clinical application in GBM.


Subject(s)
Brain Neoplasms , Cancer Vaccines , Glioblastoma , Immunotherapy , Glioblastoma/therapy , Glioblastoma/immunology , Humans , Immunotherapy/methods , Brain Neoplasms/therapy , Brain Neoplasms/immunology , Cancer Vaccines/therapeutic use , Immune Checkpoint Inhibitors/therapeutic use , Dendritic Cells/immunology , Tumor-Associated Macrophages/immunology , T-Lymphocytes/immunology
5.
Oral Oncol ; 151: 106723, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38387261

ABSTRACT

OBJECTIVE: This study was designed to assess the efficacy and safety of cadonilimab monotherapy, a first-in-class, bi-specific PD-1/CTLA-4 antibody, in patients with previously treated recurrent or metastatic nasopharyngeal carcinoma (R/M-NPC). PATIENTS AND METHODS: This multicenter, open-label, single-arm, phase II clinical trial enrolled patients with R/M-NPC who had failed first-line platinum-based chemotherapy and second-line single agent or combined chemotherapy, and immunotherapy-naive. Patients received cadonilimab for 6 mg/kg once every 2 weeks (Q2W). The primary endpoint was objective response rate (ORR) in full analysis set (FAS) assessed by investigators according to RECIST v.1.1. The secondary endpoint included progression-free survival (PFS), overall survival (OS), duration of response (DoR), time to response (TTR) and safety. RESULTS: A total of 23 patients were assessed. The median time from first dose to data cutoff was 16.56 (range, 0.8-25.2) months. ORR was 26.1 % (95 %CI:10.2-48.4). The ORR were 44.4 % (95 %CI: 13.7-78.8) and 14.3 % (95 %CI:1.8-42.8) in patients with tumor PD-L1 expression ≥50 % and <50 %, respectively. ORR was achieved in 40.0 % (95 %CI:12.2-73.8) of patients with EBV-DNA level <4000 IU/ml (n = 10) and 15.4 % (95 %CI:1.9-45.4) of those with ≥4000 IU/ml. The median PFS was 3.71 months (95 %CI: 1.84-9.30). respectively. Median OS was not reached, and the 12-month OS rate was 79.7 % (95 % CI:54.5-91.9). Only two patients (8.3 %) experienced Grade ≥3 treatment-related adverse events (TRAEs) with hypothyroidism (30.4 %), rash (21.7 %) and pruritus (21.7 %) being the most prevalent TRAEs. CONCLUSION: Cadonilimab monotherapy demonstrated a promising efficacy and manageable toxicity in patients with previously treated R-M/NPC and provide an efficacious salvage treatment option.


Subject(s)
Nasopharyngeal Neoplasms , Humans , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Nasopharyngeal Carcinoma/drug therapy , Nasopharyngeal Neoplasms/pathology , Progression-Free Survival , Treatment Outcome
6.
Clin Transl Oncol ; 26(5): 1063-1076, 2024 May.
Article in English | MEDLINE | ID: mdl-37921958

ABSTRACT

Non-small-cell lung cancer (NSCLC) has an extremely low 5-year survival rate, with the only effective treatment being immunoradiotherapy (iRT). Here, we review the progress of clinical research on iRT for non-small-cell lung cancer (NSCLC) over 2018-2023, as well as the future directions. We first discuss the synergistic mechanisms of iRT, reflected in three aspects: immune regulation of RT, RT-activated immune-related pathways, and RT-related immune sensitization. iRT may include either external-beam or stereotactic-body RT combined with either immune checkpoint inhibitors (e.g., immunoglobulins against immune programmed cell death (PD) 1/PD ligand 1 or CD8+ T lymphocyte antigen 4) or traditional Chinese medicine drugs. Regarding clinical effectiveness and safety, iRT increases overall and progression-free survival and tumor control rate among patients with NSCLC but without a considerable increase in toxicity risk. We finally discuss iRT challenges and future directions reported over 2018-2023.

7.
J Oncol Pharm Pract ; : 10781552231171881, 2023 May 10.
Article in English | MEDLINE | ID: mdl-37161281

ABSTRACT

INTRODUCTION: Checkpoint inhibitors (PCI) have reached an important place in the pharmaceutical market in the treatment of various types of cancer. However, due to immune-related adverse events (IRAE) to the treatment, patients with preexisting autoimmune diseases (PAD) are excluded from clinical studies, leading to a large gap in knowledge on this topic. This study aims to discuss the use of PCI in the patients with cancer and PAD by an integrative review. METHODS: For this integrative review we carried out research from 2013 to 2022 using database platforms for observational studies reporting data from safety and efficacy of PCI in patients with cancer and PAD. RESULTS: The search resulted in 161 articles and after applying the exclusion criteria, 15 clinical studies that adopted a retrospective observational design were selected and analyzed. The age range of patients was 54-71 years, with 19-68% male. The proportion of patients clinically active or receiving immunosuppressants who were initiated on PCI ranged from 0% to 57% and 14% to 73%, respectively. The mean reported follow-up time ranged from 8.0 to 16.8 months. The occurrence of an outbreak or the new IRAE had an average of 32.6%. CONCLUSION: IRAE are frequent in patients who use PCI and have cancer and PAD, carrying discontinuation of therapy. However, the multidisciplinary team needs to be aligned to manage these situations in the best way.

8.
Clin Transl Oncol ; 25(2): 396-407, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36138335

ABSTRACT

BACKGROUND: Wilms' tumor 1 (WT1) is highly expressed in various solid tumors and hematologic malignancies. DSP-7888 (adegramotide/nelatimotide) Emulsion is an investigational therapeutic cancer vaccine comprising three synthetic epitopes derived from WT1. We evaluated the mechanism of action of DSP-7888 Emulsion, which is hypothesized to induce WT1-specific cytotoxic T lymphocytes (CTLs) and helper T lymphocytes (HTLs). METHODS: The ability of nelatimotide and adegramotide to induce WT1-specific CD8+ T cells and CD4+ T cells was assessed in human peripheral blood mononuclear cells (PBMCs). The ability of DSP-7888 Emulsion to induce WT1-specific CTLs in vivo was assessed using human leukocyte antigen-I (HLA-I) transgenic mice. To assess how adegramotide, the helper peptide in DSP-7888 Emulsion, enhances WT1-specific CTLs, HLA-I transgenic mice were administered DSP-7888 or nelatimotide-only Emulsion. Interferon-gamma secretion under antigen stimulation by splenocytes co-cultured with or without tumor cells was then quantified. The effects of combination treatment with DSP-7888 Emulsion and an anti-programmed cell death protein 1 (PD-1) antibody on tumor volume and the frequency of tumor-infiltrating WT1-specific T cells were assessed in HLA-I transgenic mice implanted with WT1 antigen-positive tumors. RESULTS: The peptides in DSP-7888 Emulsion were shown to induce WT1-specific CTLs and HTLs in both human PBMCs and HLA-I transgenic mice. Unlike splenocytes from nelatimotide-only Emulsion-treated mice, splenocytes from DSP-7888 Emulsion-treated mice exhibited high levels of interferon-gamma secretion, including when co-cultured with tumor cells; interferon-gamma secretion was further enhanced by concomitant treatment with anti-PD-1. HLA-I transgenic mice administered DSP-7888 Emulsion plus anti-PD-1 experienced significantly greater reductions in tumor size than mice treated with either agent alone. This reduction in tumor volume was accompanied by increased numbers of tumor-infiltrating WT1-specific CTLs. CONCLUSIONS: DSP-7888 Emulsion can promote both cytotoxic and helper T-cell-mediated immune responses against WT1-positive tumors. Adegramotide enhances CTL numbers, and the CTLs induced by treatment with both nelatimotide and adegramotide are capable of functioning within the immunosuppressive tumor microenvironment. The ability of anti-PD-1 to enhance the antitumor activity of DSP-7888 Emulsion in mice implanted with WT1-positive tumors suggests the potential for synergy.


Subject(s)
Cancer Vaccines , Neoplasms , Humans , Mice , Animals , CD8-Positive T-Lymphocytes , Cancer Vaccines/therapeutic use , Interferon-gamma/metabolism , Emulsions/metabolism , Emulsions/pharmacology , Leukocytes, Mononuclear/metabolism , WT1 Proteins , Neoplasms/metabolism , Peptides , T-Lymphocytes, Cytotoxic , Mice, Transgenic , Tumor Microenvironment
9.
Clin Transl Oncol ; 25(4): 1067-1079, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36512305

ABSTRACT

PURPOSE: The interaction between tumor cells and immune system in hepatocellular carcinoma (HCC) remains unclear. Great clinical achievements have progressed in HCC patients treated with immune checkpoint inhibitors (ICIs) for programmed death-1 and its ligands. However, response efficacy for these therapies is limited, thereby requiring alternative ICI candidates for HCC treatment. B7 homolog 3 protein (B7-H3), an immunoregulatory protein, plays a significant role in tumor immunity and disease progression. In this study, we evaluated the correlation between B7-H3 expression and prognosis of HCC patients, and investigated the therapeutic potential of B7-H3 targeting in HCC. METHODS: B7-H3 expression was analyzed immunohistochemically in HCC patients, and its relationship with tumor-infiltrating lymphocyte infiltration was assessed. The anti-tumor efficacy of anti-B7-H3 antibody therapy was determined using an in vitro co-culture system and a subcutaneous HCC-bearing murine model. RESULTS: We found that B7-H3 overexpressed in tumor cells and positively correlated with poor prognosis in HCC patients. B7-H3 inhibited the infiltration of CD8+ T cells in tumors. Furthermore, co-culture experiment indicated that inhibiting B7-H3 in tumor cells significantly increased T cells-mediated immune activities and tumor cell killing. Consistently, anti-B7-H3 antibody-treated HCC murine model showed decreased tumor size and enhanced anti-tumor immunity mediated by CD8+ T cells. CONCLUSION: Altogether, our findings suggest that B7-H3 inhibition in tumor cells restores the immune cytotoxicity of T cells, which in turn promotes apoptosis of target cells. Therefore, B7-H3 serves as a key negative regulator in tumor immunity and the promising clinical utility of B7-H3-based immunotherapies for HCC treatment could be developed.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Animals , Mice , Carcinoma, Hepatocellular/metabolism , CD8-Positive T-Lymphocytes , Liver Neoplasms/metabolism , Disease Models, Animal , B7 Antigens/metabolism , Transcription Factors/metabolism
10.
Ecancermedicalscience ; 17: 1643, 2023.
Article in English | MEDLINE | ID: mdl-38414972

ABSTRACT

In Colombia, renal cancer is a rare condition, with clear cell renal cell carcinoma (ccRCC) being the most prevalent neoplasm. In recent years, immune checkpoint inhibitors (ICI) have been proposed for the management of metastatic disease, as they have shown improved rates of response and long-term survival. Furthermore, they exhibit a favourable tolerance profile, and adverse events causing significant morbidity are infrequent. We report the case of a 61-year-old male patient initially diagnosed with early-stage ccRCC who underwent right nephrectomy in 2009. Six years later, disease recurrence with metastatic compromise was documented, which led to the resection of the L1 vertebral body followed by radiotherapy and maintenance treatment with sunitinib. Due to disease progression, treatment with sunitinib was discontinued. Subsequently, everolimus was initiated as second-line immunotherapy, which was later discontinued due to the appearance of new metastatic lesions. In 2017, the patient was referred to our institution, where a third-line pharmacological treatment with nivolumab was initiated. In 2022, complete remission by positron emission tomography-computed tomography (PET-CT) was evidenced, which has been sustained to date. This case demonstrates the efficacy and safety of ICI in patients with metastatic ccRCC. The case presented is relevant in that it describes the achievement of complete remission in a patient who did not respond to the first two lines of immunotherapy. Given the limited literature regarding the discontinuation of therapy after achieving sustained remission, further research is warranted to explore this topic.

11.
Arq. bras. oftalmol ; Arq. bras. oftalmol;86(5): e2023, 2023. graf
Article in English | LILACS-Express | LILACS | ID: biblio-1527803

ABSTRACT

ABSTRACT Iridociliary ring melanoma is an uncommon type of uveal melanoma. Clinical manifestation varies from asymptomatic cases to masquerade syndromes mimicking refractory glaucoma. Treatment options include radiotherapy and enucleation. Management of metastatic uveal melanoma remains discouraging. Novel therapies using immune checkpoint inhibitors are currently under study. We present a case of a 54-year-old Hispanic woman with progressive vision loss due to metastatic ring melanoma with anterior chamber seeding treated with pembrolizumab.


RESUMO O melanoma iridociliar em anel é um tipo incomum de melanoma uveal. As manifestações clínicas variam desde casos assintomáticos até síndromes mascaradas que mimetizam um glaucoma refratário. As opções de tratamento incluem radioterapia e enucleação. O manejo do melanoma uveal metastático continua desanimador. Novas terapias usando inibidores de checkpoint imunológico estão atualmente em estudo. Apresentamos o caso de uma mulher hispânica de 54 anos com perda progressiva da visão por um melanoma metastático em anel, com semeadura de câmara anterior, tratada com pembrolizumabe.

12.
Front Immunol ; 13: 984349, 2022.
Article in English | MEDLINE | ID: mdl-36091058

ABSTRACT

Lung cancer is the second most common and the most lethal malignancy worldwide. It is estimated that lung cancer in never smokers (LCINS) accounts for 10-25% of cases, and its incidence is increasing according to recent data, although the reasons remain unclear. If considered alone, LCINS is the 7th most common cause of cancer death. These tumors occur more commonly in younger patients and females. LCINS tend to have a better prognosis, possibly due to a higher chance of bearing an actionable driver mutation, making them amenable to targeted therapy. Notwithstanding, these tumors respond poorly to immune checkpoint inhibitors (ICI). There are several putative explanations for the poor response to immunotherapy: low immunogenicity due to low tumor mutation burden and hence low MANA (mutation-associated neo-antigen) load, constitutive PD-L1 expression in response to driver mutated protein signaling, high expression of immunosuppressive factors by tumors cells (like CD39 and TGF-beta), non-permissive immune TME (tumor microenvironment), abnormal metabolism of amino acids and glucose, and impaired TLS (Tertiary Lymphoid Structures) organization. Finally, there is an increasing concern of offering ICI as first line therapy to these patients owing to several reports of severe toxicity when TKIs (tyrosine kinase inhibitors) are administered sequentially after ICI. Understanding the biology behind the immune response against these tumors is crucial to the development of better therapeutic strategies.


Subject(s)
Lung Neoplasms , Smokers , Female , Humans , Immunologic Factors/therapeutic use , Immunotherapy , Lung Neoplasms/drug therapy , Tumor Microenvironment/genetics
13.
Front Immunol ; 13: 842555, 2022.
Article in English | MEDLINE | ID: mdl-35432383

ABSTRACT

The CSF-470 vaccine (VACCIMEL) plus BCG and GM-CSF as adjuvants has been assayed in cutaneous melanoma patients. In the adjuvant randomized Phase II study CASVAC-0401, vaccinated patients had longer distant metastasis-free survival (DMFS) than those treated with IFNα2b. Five years after locking the data, an actualization was performed. The benefit in DMFS was maintained in the vaccinated group versus the IFNα2b-treated group (p = 0.035), with a median DMFS of 96 months for VACCIMEL and 13 months for IFNα2b. The favorable risk-benefit ratio was maintained. DMFS was also analyzed as a single cohort in all the IIB, IIC, and III patients (n = 30) who had been treated with VACCIMEL. The median DMFS was 169 months, and at 48 months follow-up, it was 71.4%, which was not statistically different from DMFS of previously published results obtained in adjuvancy with ipilimumab, pembrolizumab, nivolumab, or dabrafenib/trametinib. The possible toxicity of combining VACCIMEL with anti-immune checkpoint inhibitors (ICKi) was analyzed, especially since VACCIMEL was co-adjuvated with BCG in every vaccination. A patient with in-transit metastases was studied to produce a proof of concept. During treatment with VACCIMEL, the patient developed T-cell clones reactive towards tumor-associated antigens. Three years after ending the VACCIMEL study, the patient progressed and was treated with ICKi. During ICKi treatment, the patient did not reveal any toxicity due to previous BCG treatment. When she recurred after a 4-year treatment with nivolumab, a biopsy was obtained and immunohistochemistry and RNA-seq were performed. The tumor maintained expression of tumor-associated antigens and HLA-I and immune infiltration, with immunoreactive and immunosuppressive features. VACCIMEL plus BCG and GM-CSF is an effective treatment in adjuvancy for stages IIB, IIC, and III cutaneous melanoma patients, and it is compatible with subsequent treatments with ICKi.


Subject(s)
Cancer Vaccines , Hematopoietic Stem Cell Transplantation , Melanoma , Skin Neoplasms , Adjuvants, Immunologic , Antigens, Neoplasm , BCG Vaccine , Cancer Vaccines/adverse effects , Female , Granulocyte-Macrophage Colony-Stimulating Factor , Humans , Nivolumab/therapeutic use , Melanoma, Cutaneous Malignant
14.
Braz J Otorhinolaryngol ; 88 Suppl 1: S70-S81, 2022.
Article in English | MEDLINE | ID: mdl-34045134

ABSTRACT

INTRODUCTION: Evidence of programmed death-1 inhibitors in nasopharyngeal carcinoma has been accumulated. However, previous clinical studies were basically small sample size. OBJECTIVE: This study aimed to summarize existing studies to comprehensively compare programmed death-1 inhibitors in nasopharyngeal carcinoma with or without chemotherapy. METHODS: Different databases were searched for full-text publications with a programmed death-1 inhibitor with or without chemotherapy. No study-to-study heterogeneity was detected, and fixed-effect models were applied to synthesize data. RESULTS: Seven studies were included. The mean progression-free survival duration of programmed death-1 inhibitors treatment was 4.66 months. The 6 month progression-free survival rate was 50%, however, the12 month progression-free survival rate fell to 27%. Comparing with programmed death-1 inhibitor monotherapy, the objective response rate was higher in combination therapy (pooled RR=2.90, 95% CI: 2.07-4.08). The partial response rate was higher in patients receiving programmed death-1 in association with chemotherapy (pooled RR=3.09, 95% CI: 2.15-4.46), In contrast, the progressive disease rate was lower in combination therapy group (pooled RR=0.06, 95% CI: 0.01-0.31). Stable disease condition was comparable (pooled RR=0.90, 95% CI: 0.50-1.64) with or without chemotherapy. Programmed death-1 single use or combined with chemotherapy did not influence the total adverse events occurrence (pooled RR=0.99, 95% CI: 0.93-1.05). However, combination therapy could increase the risk of serious adverse events such as anemia, thrombocytopenia, and neutropenia. CONCLUSION: The present study summarized the efficacy and safety of programmed death-1 inhibitors in nasopharyngeal carcinoma. Combination therapy showed higher anti-tumor activity except for higher risk of myelosuppression.


Subject(s)
Antineoplastic Agents, Immunological , Immunotherapy , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , Humans , Nasopharyngeal Carcinoma/drug therapy , Nasopharyngeal Neoplasms/drug therapy , Antineoplastic Agents, Immunological/therapeutic use
15.
Braz. j. otorhinolaryngol. (Impr.) ; Braz. j. otorhinolaryngol. (Impr.);88(supl.1): 70-81, 2022. tab, graf
Article in English | LILACS-Express | LILACS | ID: biblio-1420802

ABSTRACT

Abstract Introduction Evidence of programmed death-1 inhibitors in nasopharyngeal carcinoma has been accumulated. However, previous clinical studies were basically small sample size. Objective This study aimed to summarize existing studies to comprehensively compare programmed death-1 inhibitors in nasopharyngeal carcinoma with or without chemotherapy. Methods Different databases were searched for full-text publications with a programmed death-1 inhibitor with or without chemotherapy. No study-to-study heterogeneity was detected, and fixed-effect models were applied to synthesize data. Results Seven studies were included. The mean progression-free survival duration of programmed death-1 inhibitors treatment was 4.66 months. The 6 month progression-free survival rate was 50%, however, the12 month progression-free survival rate fell to 27%. Comparing with programmed death-1 inhibitor monotherapy, the objective response rate was higher in combination therapy (pooled RR = 2.90, 95% CI: 2.07-4.08). The partial response rate was higher in patients receiving programmed death-1 in association with chemotherapy (pooled RR = 3.09, 95% CI: 2.15-4.46), In contrast, the progressive disease rate was lower in combination therapy group (pooled RR = 0.06, 95% CI: 0.01-0.31). Stable disease condition was comparable (pooled RR = 0.90, 95% CI: 0.50-1.64) with or without chemotherapy. Programmed death-1 single use or combined with chemotherapy did not influence the total adverse events occurrence (pooled RR = 0.99, 95% CI: 0.93-1.05). However, combination therapy could increase the risk of serious adverse events such as anemia, thrombocytopenia, and neutropenia. Conclusion The present study summarized the efficacy and safety of programmed death-1 inhibitors in nasopharyngeal carcinoma. Combination therapy showed higher anti-tumor activity except for higher risk of myelosuppression.


Resumo Introdução Há um acúmulo de evidências sobre os inibidores de morte celular programada‐1 no carcinoma nasofaríngeo. Porém, os estudos clínicos anteriores foram quase todos feitos com amostras de tamanho pequeno. Objetivo Resumir os estudos existentes para comparar de forma abrangente os inibidores de morte celular programada‐1 em carcinoma nasofaríngeo com ou sem quimioterapia. Método A pesquisa foi feita em diferentes bases de dados em busca de publicações de texto completo que usaram um inibidor de morte celular programada‐1 com ou sem quimioterapia. Nenhuma heterogeneidade entre os estudos foi detectada e modelos de efeito fixo foram aplicados para sintetizar os dados. Resultados Sete estudos foram incluídos. A duração média da sobrevida livre de progressão no tratamento com inibidores de morte celular programada‐1 foi de 4,66 meses. A taxa de sobrevida livre de progressão em seis meses foi de 50%; no entanto, a taxa de sobrevida livre de progressão em 12 meses caiu para 27%. Em comparação com a monoterapia com inibidor de morte celular programada‐1, a taxa de resposta objetiva (taxa de resposta combinada = 2,90, IC de 95%: 2,07-4,08). A taxa de resposta parcial foi maior em pacientes que receberam morte celular programada‐1 em combinação com quimioterapia (taxa de resposta combinada = 3,09, IC 95%: 2,15‐4,46). Ao contrário, a taxa de progressão da doença foi menor no grupo com terapia combinada (taxa de resposta combinada = 0,06, IC de 95%: 0,01-0,31). A condição de estabilidade da doença com ou sem quimioterapia foi comparável (taxa de resposta combinada = 0,90, IC de 95%: 0,50-1,64). O uso isolado de morte celular programada‐1 ou combinado com quimioterapia não influenciou a ocorrência de eventos adversos totais (taxa de resposta combinada = 0,99, IC de 95%: 0,93-1,05). No entanto, a terapia combinada pode aumentar o risco de eventos adversos graves, como anemia, trombocitopenia e neutropenia. Conclusão O presente estudo fez um resumo da eficácia e segurança dos inibidores de morte celular programada‐1 em carcinoma nasofaríngeo. A terapia combinada mostrou uma atividade antitumoral maior, excetuando‐se o risco maior de mielossupressão.

16.
Medwave ; 21(5): e8202, 2021 Jun 04.
Article in Spanish, English | MEDLINE | ID: mdl-34214067

ABSTRACT

In the last decade, the development of immune checkpoint inhibitors have revolutionized the treatment of patients with advanced renal cell carcinoma, with the potential for dramatic changes in the therapeutic landscape. Nivolumab, a monoclonal antibody inhibitor of transmem-brane programmed cell death protein 1 (PD-1), was approved as monotherapy in 2015 for advanced renal cell carcinoma in patients previously treated with an agent targeting vascular endothelial growth factor. In April 2018, the combination of nivolumab and ipilimumab, a cytotoxic T-lymphocyte-associated antigen 4 inhibitor, was approved for patients with previously untreated intermediate- and poor-risk advanced renal cell carcinoma. Then, in 2019, combination therapies consisting of pembrolizumab (anti-PD-1) or avelumab (anti-PD-1 ligand, PD-L1) with axitinib (a vascular endothelial growth factor receptor tyrosine kinase inhibitor) were also approved for use in all risk groups. This review pre-sents a brief historical review of the association between immunology and oncology; describes essential aspects of the mechanism of action of immune checkpoint inhibitors; discusses the current evidence regarding the clinical use of different immunotherapy regimens for the treatment of patients with renal cell carcinoma, both clear cell and other histological types; and provides general information on their adverse effects. The role of appropriate patient selection is analyzed to allow individualization of therapy and improve the already promising results. Finally, per-spectives on the future use of immune checkpoint inhibitors to treat renal cancer are discussed.


En la última década, el desarrollo de inhibidores de puntos de control o checkpoints inmunológicos, ha revolucionado el tratamiento de los pacientes con carcinoma de células renales avanzado avizorándose posibles cambios dramáticos en el escenario terapéutico. Nivolumab, un anticuerpo monoclonal inhibidor de la proteína transmembrana de muerte celular programada 1 (PD-1), se aprobó como monoterapia en 2015 para carcinoma de células renales avanzado en pacientes previamente tratados con algún agente dirigido al factor de crecimiento endotelial vascular. En abril de 2018, la combinación de nivolumab e ipilimumab, un inhibidor del CTLA-4, fue aprobado para pacientes con carcinoma de células renales avanzado de riesgo intermedio y riesgo desfavorable, previamente no tratados. Luego, en 2019, terapias combinadas que consisten en pembrolizumab (anti-PD-1) o avelumab (anti-ligando-PD-1, PD-L1) con axitinib (un inhibidor del receptor tirosina kinasa del factor de crecimiento endotelial vascular); también fueron aprobadas para su uso en todos los grupos de riesgo. En esta revisión se presenta una breve reseña histórica sobre la asociación entre la inmunología y la oncología; se describen aspectos básicos del mecanismo de acción de los inhibidores de puntos de control inmunológicos; se discute la evidencia actual relacionada con el uso clínico de los distintos esquemas de inmu-noterapia para el tratamiento de pacientes con carcinoma de células renales, tanto de células claras como de otros tipos histológicos; y se entrega información general sobre sus efectos adversos. Se analiza el rol de la adecuada selección de pacientes que permita una individualización de la terapia y, por ende, una mejora de los ya promisorios resultados. Por último, se discuten las perspectivas sobre el uso futuro de los inhibidores de puntos de control inmunológicos para el tratamiento del cáncer renal.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Carcinoma, Renal Cell/therapy , Immunotherapy/methods , Kidney Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , CTLA-4 Antigen/antagonists & inhibitors , Carcinoma, Renal Cell/drug therapy , Humans , Immune Checkpoint Inhibitors , Nivolumab/therapeutic use , Programmed Cell Death 1 Receptor/immunology , Treatment Outcome , Vascular Endothelial Growth Factor A
17.
Ther Adv Urol ; 13: 17562872211029779, 2021.
Article in English | MEDLINE | ID: mdl-34290827

ABSTRACT

Immunotherapy, in the form of immune checkpoint inhibitors (ICI), has shown activity in metastatic urothelial bladder carcinoma, resulting in the approval of several ICI agents in the first- and second-line settings. This has led to an increased interest in studying their efficacy in the neoadjuvant setting for muscle invasive disease - an area of significant unmet need. This non-systematic review will look at the evidence supporting the use of ICI in the neoadjuvant setting for this tumor, results of early-phase studies, ongoing trials, and possible future applications for these drugs.

18.
Clin Transl Oncol ; 23(9): 1818-1826, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33728869

ABSTRACT

INTRODUCTION: The efficacy of immune checkpoint inhibitors in patients with brain metastases (BMs) from non-oncogene addicted non-small cell lung cancer (NSCLC) is under investigation. Here, we sought to determine the optimal management of NSCLCs with PD-L1 ≥ 50% and asymptomatic BMs who were treated with first-line pembrolizumab. METHODS: Thirty patients from 15 institutions with PD-L1 ≥ 50% NSCLC had asymptomatic BMs, and met inclusion criteria. Patients were classified based on whether they had undergone upfront local radiotherapy for BMs as well as on the type of brain radiotherapy received. RESULTS: Nine patients were treated with upfront pembrolizumab alone, 8 patients with whole-brain radiotherapy (WBRT) followed by pembrolizumab and 13 patients with stereotactic radiosurgery (SRS) followed by pembrolizumab. Patients' characteristics were similar among the three groups of patients except for a higher number of BMs ≥ 3 in the WBRT group. One complete and 4 partial intracranial responses were observed with upfront pembrolizumab alone. The median survival was not reached for the pembrolizumab and WBRT (n = 8) groups, and it was 7.6 months for the SRS (n = 13) group (P = 0.09), with 12-month survival rates being 55.5%, 62.5%, and 23.0%, respectively. Salvage WBRT was delivered in 1 patient in the upfront pembrolizumab group and in 4 patients in the SRS group. CONCLUSIONS: Upfront pembrolizumab showed efficacy in selected patients with PD-L1 ≥ 50% non-oncogene addicted NSCLC and asymptomatic BMs. Prospective studies should address whether pembrolizumab alone, and deferral of radiotherapy, could be pursued in this patient population.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Asymptomatic Diseases , B7-H1 Antigen/metabolism , Brain Neoplasms/drug therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Agents, Immunological/adverse effects , Brain Neoplasms/mortality , Brain Neoplasms/radiotherapy , Brain Neoplasms/secondary , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/radiotherapy , Carcinoma, Non-Small-Cell Lung/secondary , Cranial Irradiation/methods , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Middle Aged , Radiosurgery/statistics & numerical data , Retrospective Studies , Salvage Therapy/methods , Treatment Outcome
19.
Clin Transl Oncol ; 23(8): 1630-1636, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33586122

ABSTRACT

BACKGROUND: Although the immune checkpoint inhibitors (ICIs) became a vital part of cancer care, many patients do not respond to treatment, indicating need for biomarkers. The Pan-Immune-Inflammation Value (PIV) is a recently developed peripheral blood count-based biomarker. Herein, we evaluated a PIV-based candidate scoring system as a prognostic biomarker in ICI-treated patients. METHODS: A total of 120 advanced cancer patients treated with anti-PD-1 or anti-PD-L1 inhibitors for any cancer type were included in this study. The PILE scoring system incorporating the PIV (< median vs. ≥ median), lactate dehydrogenase levels (normal vs. > normal) and Eastern Cooperative Oncology Group performance status (0 vs. ≥ 1) was constructed from the multivariate analyses and used for stratification. The association between overall survival (OS), progression-free survival and PILE risk category was evaluated with multivariate analysis. RESULTS: The median follow-up was 9.62 months and the median OS of all cohort were 12.42 ± 2.75 months. Patients with higher PIV had significantly decreased OS (7.75 ± 1.64 vs. 18.63 ± 4.26 months, p = 0.037). The patients in the PILE high-risk group (PILE score 2-3) had decreased OS (18.63 ± 4.02 vs. 5.09 ± 1.23 months, HR: 2.317, 95% CI: 1.450-3.700, p < 0.001) and PFS (7.69 ± 1.30 vs. 2.69 ± 0.65 months, HR: 1.931, 95% CI: 1.263-2.954, p = 0.002) compared to PILE low-risk group (PILE score 0-1). The Harrell C-Index values were 0.65 and 0.61 for OS and PFS prediction, respectively. CONCLUSION: In this study, we demonstrated a decreased overall survival in ICI-treated patients with a higher PILE score. If prospective studies validate our results, PILE score could be a biomarker for immunotherapy.


Subject(s)
Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Neoplasms/therapy , Biomarkers , Blood Cell Count , Female , Humans , Inflammation/blood , Inflammation/mortality , L-Lactate Dehydrogenase/blood , Male , Multivariate Analysis , Neoplasms/blood , Neoplasms/mortality , Prognosis , Progression-Free Survival , Sensitivity and Specificity , Severity of Illness Index
20.
Cancers (Basel) ; 13(3)2021 Feb 02.
Article in English | MEDLINE | ID: mdl-33540543

ABSTRACT

Tumor necrosis factor alpha (TNFα) is a pleiotropic cytokine known to have contradictory roles in oncoimmunology. Indeed, TNFα has a central role in the onset of the immune response, inducing both activation and the effector function of macrophages, dendritic cells, natural killer (NK) cells, and B and T lymphocytes. Within the tumor microenvironment, however, TNFα is one of the main mediators of cancer-related inflammation. It is involved in the recruitment and differentiation of immune suppressor cells, leading to evasion of tumor immune surveillance. These characteristics turn TNFα into an attractive target to overcome therapy resistance and tackle cancer. This review focuses on the diverse molecular mechanisms that place TNFα as a source of resistance to immunotherapy such as monoclonal antibodies against cancer cells or immune checkpoints and adoptive cell therapy. We also expose the benefits of TNFα blocking strategies in combination with immunotherapy to improve the antitumor effect and prevent or treat adverse immune-related effects.

SELECTION OF CITATIONS
SEARCH DETAIL