Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Semin Liver Dis ; 42(2): 212-224, 2022 05.
Article in English | MEDLINE | ID: mdl-35263795

ABSTRACT

The complex immune system of the liver has a major role in tumor surveillance, but also partly explains why current immune therapies are poorly effective against liver cancers. Known primarily for its tolerogenic capacity, the hepatic immune repertoire also comprises diverse populations of armored immune cells with tumor surveillant roles. In healthy people, these work together to successfully identify malignant cells and prevent their proliferation, thus halting tumor formation. When frontline hepatic immune surveillance systems fail, compromised hepatic immunity, driven by obesity, infection, or other pathological factors, allows primary or secondary liver cancers to develop. Tumor growth promotes the normal tolerogenic immunological milieu of the liver, perhaps explaining why current immunotherapies fail to work. This review explores the complex local liver immune system with the hope of identifying potential therapeutic targets needed to best overcome immunological barriers in the liver to create an environment no longer hostile to immunotherapy for the treatment of liver cancer.


Subject(s)
Immunotherapy , Liver Neoplasms , Humans , Liver Neoplasms/therapy , Tumor Microenvironment
2.
Oncologist ; 27(3): e273-e285, 2022 03 11.
Article in English | MEDLINE | ID: mdl-35274717

ABSTRACT

BACKGROUND: We conducted a phase II study of the combination of pembrolizumab with capecitabine and oxaliplatin (CAPOX) in patients with advanced biliary tract carcinoma (BTC) to assess response rate and clinical efficacy. Exploratory objectives included correlative studies of immune marker expression, tumor evolution, and immune infiltration in response to treatment. PATIENTS AND METHODS: Adult patients with histologically confirmed BTC were enrolled and received oxaliplatin and pembrolizumab on day 1 of cycles 1-6. Capecitabine was administered orally twice daily as intermittent treatment, with the first dose on day 1 and the last dose on day 14 of cycles 1-6. Starting on cycle 7, pembrolizumab monotherapy was continued until disease progression. The primary endpoint was progression-free survival (PFS). Secondary endpoints were safety, tolerability, feasibility, and response rate. Immunohistochemistry (IHC) for PD-L1 and immune infiltrates was analyzed in paired tumor biopsies, as well as bulk transcriptome and exome profiling for five patients and single-cell RNA sequencing for one partial responder. RESULTS: Eleven patients enrolled, three of whom had received no prior systemic therapy. Treatment was well tolerated, and the most common treatment-related grade 3 or 4 adverse events were lymphocytopenia, anemia, and decreased platelet count. Three patients (27.3%) achieved a partial response, and six (54%) had stable disease. The disease control rate was 81.8%. The median PFS was 4.1 months with a 6-month PFS rate of 45.5%. Molecular profiling suggests qualitative differences in immune infiltration and clonal evolution based on response. CONCLUSION: Capecitabine and oxaliplatin in combination with pembrolizumab is tolerable and a potentially effective treatment for refractory advanced BTC. This study highlights a design framework for the precise characterization of individual BTC tumors. TRIAL REGISTRATION: This study was registered in ClinicalTrials.gov (NCT03111732).


Subject(s)
Bile Duct Neoplasms , Biliary Tract , Carcinoma , Gastrointestinal Neoplasms , Adult , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bile Duct Neoplasms/drug therapy , Capecitabine/adverse effects , Carcinoma/drug therapy , Gastrointestinal Neoplasms/drug therapy , Humans , Oxaliplatin
3.
Lancet ; 403(10442): 2370-2371, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824947
4.
Hepatology ; 69(5): 2048-2060, 2019 05.
Article in English | MEDLINE | ID: mdl-30578687

ABSTRACT

Treatment options for patients with advanced biliary tract cancer are limited. Dysregulation of the immune system plays an important role in the pathogenesis of biliary tract cancer (BTC). This study aimed to investigate whether tremelimumab, an anti-CTLA4 (cytotoxic T-lymphocyte-associated protein 4) inhibitor, could be combined safely with microwave ablation to enhance the effect of anti-CTLA4 treatment in patients with advanced BTC. Patients were enrolled to receive monthly tremelimumab (10 mg/kg, intravenously) for six doses, followed by infusions every 3 months until off-treatment criteria were met. Thirty-six days after the first tremelimumab dose, patients underwent subtotal microwave ablation. Interval imaging studies were performed every 8 weeks. Adverse events (AEs) were noted and managed. Tumor and peripheral blood samples were collected to perform immune monitoring and whole-exome sequencing (WES). Twenty patients with refractory BTC were enrolled (median age, 56.5 years). No dose-limiting toxicities were encountered. The common treatment-related AEs included lymphopenia, diarrhea, and elevated transaminases. Among 16 patients evaluable for efficacy analysis, 2 (12.5%) patients achieved a confirmed partial response (lasting for 8.0 and 18.1 months, respectively) and 5 patients (31.3%) achieved stable disease. Median progression free survival (PFS) and overall survival (OS) were 3.4 months (95% confidence interval [CI], 2.5-5.2) and 6.0 months (95% CI, 3.8-8.8), respectively. Peripheral blood immune cell subset profiling showed increased circulating activated human leukocyte antigen, DR isotype ([HLA-DR] positive) CD8+ T cells. T-cell receptor (TCR)ß screening showed tremelimumab expanded TCR repertoire, but not reaching statistical significance (P = 0.057). Conclusion: Tremelimumab in combination with tumor ablation is a potential treatment strategy for patients with advanced BTC. Increased circulating activated CD8+ T cells and TCR repertoire expansion induced by tremelimumab may contribute to treatment benefit.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents/administration & dosage , Biliary Tract Neoplasms/drug therapy , Carcinoma/drug therapy , Microwaves/therapeutic use , Adult , Aged , Biliary Tract Neoplasms/immunology , Carcinoma/immunology , Female , Humans , Male , Middle Aged , Radiofrequency Therapy , Treatment Outcome
5.
Cancer Immunol Immunother ; 68(4): 599-608, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30688989

ABSTRACT

BACKGROUND: Checkpoint inhibitors have recently been approved for the treatment of patients with hepatocellular carcinoma (HCC). However, biomarkers, which will help identify patients responding to therapy, are missing. We recently tested the combination of anti-CTLA4 treatment (tremelimumab) with loco-regional therapy in patients with HCC and reported a partial response rate of 26%. METHODS: Here, we report updated survival analyses and results from our immune monitoring studies on peripheral blood mononuclear cells (PBMCs) and tumors from these patients. RESULTS: Tremelimumab therapy increased CD4+-HLA-DR+, CD4+PD-1+, CD8+HLA-DR+, CD8+PD-1+, CD4+ICOS+ and CD8+ICOS+ T cells in the peripheral blood of the treated patients. Patients with higher CD4+PD1+ cell frequency at baseline were more likely to respond to tremelimumab therapy. PD-1 expression was increased on alpha fetal protein (AFP) and survivin-specific CD8 T cells upon tremelimumab treatment. An increase of tumor infiltrating CD3+ T cells were also seen in these patients. Immunosequencing of longitudinal PBMC showed that one cycle of tremelimumab significantly decreased peripheral clonality, while no additional effects were seen after loco-regional therapy. CONCLUSION: In summary, we observed a clear activation of T cell responses in HCC patients treated with tremelimumab and identified potential biomarkers which will help identify patients responding to immunotherapy with anti-CTLA4.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , CTLA-4 Antigen/antagonists & inhibitors , Carcinoma, Hepatocellular/immunology , Liver Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Adult , Aged , Biomarkers , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cytotoxicity, Immunologic , Female , Genes, T-Cell Receptor beta , Humans , Immunophenotyping , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/metabolism , Male , Middle Aged , Neoplasm Staging , Pilot Projects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
6.
J Hepatol ; 66(3): 545-551, 2017 03.
Article in English | MEDLINE | ID: mdl-27816492

ABSTRACT

BACKGROUND & AIMS: Tremelimumab is a fully human monoclonal antibody that binds to cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) on the surface of activated T lymphocytes. Ablative therapies induce a peripheral immune response which may enhance the effect of anti-CTLA4 treatment in patients with advanced hepatocellular carcinoma (HCC). This study aimed to demonstrate whether tremelimumab could be combined safely and feasibly with ablation. METHODS: Thirty-two patients with HCC were enrolled: male:female: 28:4; median age: 62 (range 36-76). Patients were given tremelimumab at two dose levels (3.5 and 10mg/kg i.v.) every 4weeks for 6 doses, followed by 3-monthly infusions until off-treatment criteria were met. On day 36, patients underwent subtotal radiofrequency ablation or chemoablation. Staging was performed by contrast-enhanced CT or MRI scan every 8weeks. RESULTS: No dose-limiting toxicities were encountered. The most common toxicity was pruritus. Of the 19 evaluable patients, five (26.3%; 95% CI: 9.1-51.2%) achieved a confirmed partial response. Twelve of 14 patients with quantifiable HCV experienced a marked reduction in viral load. Six-week tumor biopsies showed a clear increase in CD8+ T cells in patients showing a clinical benefit only. Six and 12-month probabilities of tumor progression free survival for this refractory HCC population were 57.1% and 33.1% respectively, with median time to tumor progression of 7.4months (95% CI 4.7 to 19.4months). Median overall survival was 12.3months (95% CI 9.3 to 15.4months). CONCLUSIONS: Tremelimumab in combination with tumor ablation is a potential new treatment for patients with advanced HCC, and leads to the accumulation of intratumoral CD8+ T cells. Positive clinical activity was seen, with a possible surrogate reduction in HCV viral load. LAY SUMMARY: Studies have shown that the killing of tumors by direct methods (known as ablation) can result in the immune system being activated or switched on. The immune system could potentially also recognize and kill the cancer that is left behind. There are new drugs available known as immune checkpoint inhibitors which could enhance this effect. Here, we test one of these drugs (tremelimumab) together with ablation. CLINICAL TRIAL NUMBER: ClinicalTrials.gov: NCT01853618.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Carcinoma, Hepatocellular/therapy , Liver Neoplasms/therapy , Ablation Techniques , Adult , Aged , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/therapeutic use , CTLA-4 Antigen/antagonists & inhibitors , Carcinoma, Hepatocellular/immunology , Combined Modality Therapy , Female , Humans , Liver Neoplasms/immunology , Male , Middle Aged , Pilot Projects
7.
Hepatology ; 64(5): 1785-1791, 2016 11.
Article in English | MEDLINE | ID: mdl-27177447

ABSTRACT

Biliary tract cancers (BTC) comprise a group of uncommon malignancies in which the standard therapies are minimally effective and evolve slowly. Like the majority of gastrointestinal cancers, with some notable exceptions, the impact of immune-based approaches has yet to be seen. However, the etiological background of BTC-overlapping in almost every known causative or associated factor with inflammation-provides a strong clue that these approaches may have an impact in this group of diseases. This review covers what we currently know about the role of the immune system in the etiology of BTC, highlighting differences by subtype, and pointing to the therapeutic opportunities currently entering the clinic or about to do so. (Hepatology 2016;64:1785-1791).


Subject(s)
Biliary Tract Neoplasms/immunology , Biliary Tract Neoplasms/therapy , Humans , Immunotherapy/methods , Translational Research, Biomedical
8.
Cancer ; 122(11): 1757-65, 2016 06 01.
Article in English | MEDLINE | ID: mdl-26998818

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) incidence has been increasing in the United States for several decades; and, as the incidence of hepatitis C virus (HCV) infection declines and the prevalence of metabolic disorders rises, the proportion of HCC attributable to various risk factors may be changing. METHODS: Data from the Surveillance, Epidemiology, and End Results-Medicare linkage were used to calculate population attributable fractions (PAFs) for each risk factor over time. Patients with HCC (n = 10,708) who were diagnosed during the years 2000 through 2011 were compared with a 5% random sample of cancer-free controls (n = 332,107) residing in the Surveillance, Epidemiology, and End Results areas. Adjusted odds ratios (ORs) and PAFs were calculated for HCV, hepatitis B virus (HBV), metabolic disorders, alcohol-related disorders, smoking, and genetic disorders. RESULTS: Overall, the PAF was greatest for metabolic disorders (32%), followed by HCV (20.5%), alcohol (13.4%), smoking (9%), HBV (4.3%), and genetic disorders (1.5%). The PAF for all factors combined was 59.5%. PAFs differed by race/ethnicity and sex. Metabolic disorders had the largest PAF among Hispanics (PAF, 39.3%; 95% confidence interval [CI], 31.9%-46.7%) and whites (PAF, 34.8%; 95% CI, 33.1%-36.5%), whereas HCV had the largest PAF among blacks (PAF, 36.1%; 95% CI, 31.8%-40.4%) and Asians (PAF, 29.7%; 95% CI, 25.9%-33.4%). Between 2000 and 2011, the PAF of metabolic disorders increased from 25.8% (95% CI, 22.8%-28.9%) to 36% (95% CI, 33.6%-38.5%). In contrast, the PAFs of alcohol-related disorders and HCV remained stable. CONCLUSIONS: Among US Medicare recipients, metabolic disorders contribute more to the burden of HCC than any other risk factor, and the fraction of HCC caused by metabolic disorders has increased in the last decade. Cancer 2016;122:1757-65. Published 2016. This article is a U.S. Government work and is in the public domain in the USA..


Subject(s)
Carcinoma, Hepatocellular/etiology , Liver Neoplasms/etiology , Aged , Alcohol Drinking/adverse effects , Carcinoma, Hepatocellular/epidemiology , Case-Control Studies , Ethnicity , Female , Genetic Diseases, Inborn/complications , Hepatitis B/complications , Hepatitis C/complications , Humans , Liver Neoplasms/epidemiology , Male , Metabolic Diseases/complications , Odds Ratio , Risk Factors , SEER Program , Sex Factors , Smoking/adverse effects
9.
Invest New Drugs ; 34(2): 168-75, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26666244

ABSTRACT

BACKGROUND: KRAS mutations are clinically important predictors of resistance to EGFR-directed therapies in colorectal cancer (CRC). Oncogenic activation of the RAS/RAF/MEK/ERK signaling cascade mediates proliferation independent of growth factor signaling. We hypothesized that targeting MEK with selumetinib could overcome resistance to cetuximab in KRAS mutant CRC. METHODS: A phase I study (NCT01287130) was undertaken to determine the tolerability, and pharmacokinetic profiles of the combination of selumetinib and cetuximab, with an expanded cohort in KRAS-mutant CRC. RESULTS: 15 patients were treated in the dose escalation cohort and 18 patients were treated in the expansion cohort. Two dose-limiting toxicities were observed. One grade 3 acneiform rash and one grade 4 hypomagnesemia occurred. The most common grade 1 and 2 adverse events included rash, nausea/vomiting, diarrhea, and fatigue. The maximum tolerated dose was established at selumetinib 75 mg p.o. BID and cetuximab 250 mg/m(2) weekly following a 400 mg/m(2) load. Best clinical response in the dose escalation group included 1 unconfirmed partial response in a patient with CRC and stable disease (SD) in 5 patients (1 squamous cell carcinoma of the tonsil, 1 non-small cell lung cancer, and 3 CRC), and in the KRAS-mutant CRC dose expansion cohort, of the 14 patients who were evaluable for response, 5 patients had SD and 9 patients had progressive disease. CONCLUSIONS: The combination of selumetinib and cetuximab is safe and well tolerated. Minimal anti-tumor activity was observed in KRAS-mutant refractory metastatic CRC. Further investigations might be warranted in other cancer subtypes.


Subject(s)
Benzimidazoles/therapeutic use , Cetuximab/therapeutic use , Colorectal Neoplasms/drug therapy , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mutation/genetics , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins p21(ras)/genetics , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzimidazoles/adverse effects , Benzimidazoles/pharmacokinetics , Benzimidazoles/pharmacology , Cetuximab/adverse effects , Cetuximab/pharmacology , Colorectal Neoplasms/blood , Colorectal Neoplasms/genetics , Dose-Response Relationship, Drug , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Mitogen-Activated Protein Kinase Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Treatment Outcome
10.
J Hepatol ; 62(6): 1420-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25733155

ABSTRACT

Current systemic treatment options for patients with hepatocellular carcinoma (HCC) are limited to sorafenib. With the recent FDA approval of the second PD1-PD-L1 pathway inhibitor, immunotherapy has gained even more interest as a potential novel treatment option for patients with HCC. This is due not only because of the failure of other treatment approaches in the past, but also because immunological mechanisms have been shown to play an important role during tumor development, growth, and treatment. Here we present a review of immunological mechanisms in the liver relevant for tumor progression and treatment. We summarize our current knowledge on immune activating and immune suppressing mechanisms during tumor initiation, development, and treatment. We try to explain the paradox of how inflammatory responses in a setting of chronic infection promote tumor development, while the primary aim of immunotherapy is to activate immunity. Finally we summarize recent advances in addition to providing an outlook for the immunotherapy of HCC.


Subject(s)
Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/therapy , Immunotherapy/methods , Liver Neoplasms/immunology , Liver Neoplasms/therapy , Adoptive Transfer , Cancer Vaccines/therapeutic use , Clinical Trials as Topic , Cytokines/therapeutic use , Humans , Immune Tolerance , Immunosuppression Therapy
11.
Hepatology ; 60(5): 1637-44, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24996116

ABSTRACT

UNLABELLED: The purpose of the study was to assess the use of curative therapies for hepatocellular carcinoma (HCC) in the population. HCC treatment patterns were examined in Surveillance, Epidemiology, and End Results (SEER) 18 registries (28% of U.S.). Joinpoint regression analyses were performed to assess 2000-2010 incidence trends by tumor size, count, and receipt of potentially curative treatments (transplantation, resection, and ablation). SEER-Medicare data enabled evaluation of treatment patterns including receipt of sorafenib or transarterial chemoembolization (TACE) by HCC-associated comorbidities. Diagnoses of tumors≤5.0 cm in diameter significantly increased during 2000-2010, surpassing diagnosis of larger tumors. Overall, 23% of cases received potentially curative treatment. Joinpoint models indicated incidence rates of treatment with curative intent increased 17.6% per year during 2000-2005, then declined by -2.9% per year during 2005-2010 (P<0.001). Among HCC cases with a single tumor≤5.0 cm and no extension beyond the liver, use of ablative therapy significantly increased during 2000-2010. Use of invasive surgery for single tumors, regardless of size, significantly increased during the initial years of the decade, then plateaued. The group most likely to receive curative treatment in the SEER-Medicare cases was patients with one, small tumor confined to the liver (657 of 1,597 cases, 41%), with no difference in treatment by hepatic comorbidity status (P=0.24). A higher proportion of cases with reported liver-associated comorbidities were, however, diagnosed with tumors≤5.0 cm in diameter (1,745 0f 2,464, 71%) compared to patients with no reported comorbidities (996 of 2,596, 38%, P<0.001). CONCLUSION: Although more HCC patients were diagnosed with early disease over time, the use of curative treatments in this patient group has recently plateaued. Efforts to identify and treat more eligible candidates for curative therapy could be beneficial.


Subject(s)
Carcinoma, Hepatocellular/epidemiology , Liver Neoplasms/epidemiology , Adolescent , Adult , Aged , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Chemoembolization, Therapeutic , Child , Child, Preschool , Comorbidity , Early Diagnosis , Female , Humans , Incidence , Infant , Liver/pathology , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Male , Middle Aged , Niacinamide/analogs & derivatives , Niacinamide/therapeutic use , Phenylurea Compounds/therapeutic use , SEER Program , Sorafenib , United States/epidemiology , Young Adult
12.
Hepatology ; 57(3): 1068-77, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23112096

ABSTRACT

UNLABELLED: The presence of cirrhosis increases the potential risk of hemorrhage for patients with hepatocellular carcinoma (HCC). We evaluated the relative risk for hemorrhage in patients with HCC treated with antiangiogenic agents. We performed a systematic review and meta-analysis of antiangiogenic studies in HCC from 1995 to 2011. For nonrandomized studies we compared bleeding risk with other HCC single-arm studies that did not include an antiangiogenic agent. To separate disease-specific factors we also performed a comparison analysis with renal cell cancer (RCC)) studies that evaluated sorafenib. Sorafenib was associated with increased bleeding risk compared to control for all grade bleeding events (odds ratio [OR] 1.77; 95% confidence interval [CI] 1.04, 3.0) but not grade 3-5 events in both HCC and RCC (OR 1.46; 95% CI 0.9, 2.36; P=0.45). When comparing the risk of bleeding in single-arm phase 2 studies evaluating antiangiogenic agents, this risk for all events (OR 4.34; 95% CI 2.16, 8.73) was increased compared to control. CONCLUSION: This analysis of both randomized and nonrandomized studies evaluating an antiangiogenic agent in HCC showed that whereas the use of sorafenib was associated with an increased risk of bleeding in HCC, this was primarily for lower-grade events and similar in magnitude to the risk encountered in RCC.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/epidemiology , Hemorrhage/epidemiology , Liver Neoplasms/drug therapy , Liver Neoplasms/epidemiology , Antineoplastic Agents/therapeutic use , Humans , Incidence , Niacinamide/analogs & derivatives , Niacinamide/therapeutic use , Phenylurea Compounds/therapeutic use , Risk Factors , Sorafenib
13.
Cancer Immunol Immunother ; 62(2): 299-307, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23011590

ABSTRACT

Myeloid-derived suppressor cells (MDSC) are a heterogenous population of cells comprising myeloid progenitor cells and immature myeloid cells, which have the ability to suppress the effector immune response. In humans, MDSC have not been well characterized owing to the lack of specific markers, although it is possible to broadly classify the MDSC phenotypes described in the literature as being predominantly granulocytic (expressing markers such as CD15, CD66, CD33) or monocytic (expressing CD14). In this study, we set out to perform a direct comparative analysis across both granulocytic and monocytic MDSC subsets in terms of their frequency, absolute number, and function in the peripheral blood of patients with advanced GI cancer. We also set out to determine the optimal method of sample processing given that this is an additional source of heterogeneity. Our findings demonstrate consistent changes across sample processing methods for monocytic MDSC, suggesting that reliance upon cryopreserved PBMC is acceptable. Although we did not see an increase in the population of granulocytic MDSC, these cells were found to be more suppressive than their monocytic counterparts.


Subject(s)
Gastrointestinal Neoplasms/immunology , Granulocytes/immunology , Monocytes/immunology , Myeloid Cells/immunology , T-Lymphocytes, Regulatory/immunology , Female , Humans , Leukocytes, Mononuclear/immunology , Male , Middle Aged , Severity of Illness Index , Tumor Cells, Cultured
14.
J Vasc Interv Radiol ; 24(8): 1083-92; quiz 1093, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23885909

ABSTRACT

As personalized medicine becomes more applicable to oncologic practice, image-guided biopsies will be integral for enabling predictive and pharmacodynamic molecular pathology. Interventional radiology has a key role in defining patient-specific management. Advances in diagnostic techniques, genomics, and proteomics enable a window into subcellular mechanisms driving hyperproliferation, metastatic capabilities, and tumor angiogenesis. A new era of personalized medicine has evolved whereby clinical decisions are adjusted according to a patient's molecular profile. Several mutations and key markers already have been introduced into standard oncologic practice. A broader understanding of personalized oncology will help interventionalists play a greater role in therapy selection and discovery.


Subject(s)
Antineoplastic Agents/therapeutic use , Medical Oncology/methods , Neoplasms/drug therapy , Precision Medicine , Radiography, Interventional , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Genetic Predisposition to Disease , Genetic Testing , Genomics , Humans , Image-Guided Biopsy , Molecular Targeted Therapy , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Patient Selection , Phenotype , Predictive Value of Tests , Signal Transduction/drug effects , Signal Transduction/genetics
15.
Pharmaceuticals (Basel) ; 16(1)2023 Jan 10.
Article in English | MEDLINE | ID: mdl-36678598

ABSTRACT

In the West, recent decades have demonstrated an epidemiological trend towards esophago-gastric adenocarcinomas (EGAC), with considerable associated mortality. Historically, chemotherapy has represented the sole systemic treatment option in the advanced EGAC setting, in addition to complementing the role of surgery and radiotherapy in the case of localized disease. Immune checkpoint inhibitors (ICIs) represent a novel systemic therapeutic choice and have revolutionized the management of other malignancies, including melanoma and renal cell carcinomas. This article considers the rationale for ICIs in EGAC, reviews the evidence supporting their role in the current standard of care in EGAC, and briefly considers ongoing trials and future directions for the ICI class in EGAC.

16.
Cancers (Basel) ; 15(22)2023 Nov 14.
Article in English | MEDLINE | ID: mdl-38001661

ABSTRACT

BACKGROUND: Gastroesophageal cancers (GECs) carry considerable morbidity and mortality, and demonstrate geographical histological variances in addition to molecular heterogeneity. Consequently, the immunogenicity of the different subtypes, which can predict the likelihood of immunotherapy response, can vary. Immune checkpoint inhibitor (ICI) therapy has transformed the treatment of many cancer types over the past decade but has been slower to gain a foothold in the treatment paradigm of GECs. METHODS: This article reviews the existing evidence and use approvals for immunotherapies and immune-based treatments in GECs, in the neoadjuvant, adjuvant and metastatic disease settings. The challenges of and limitations to ICI application in current clinical practice are examined. Ongoing clinical trials and future directions of research are also considered. CONCLUSION: ICI therapy has become an established treatment option within GECs, both perioperatively and in advanced disease. However, nuances in terms of its use are not yet fully understood. Ongoing research proposes to broaden the application of immunotherapies in GECs with the potential to continue to improve outcomes.

17.
Chin Clin Oncol ; 12(5): 54, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37691344

ABSTRACT

Despite considerable international heterogeneity in the incidence and histological subtypes of gastric cancer (GC), in addition to more recent epidemiological trends, chemotherapy has long represented the main systemic therapeutic option in its treatment. For the roughly 20% of GC with human epidermal growth factor receptor 2 (HER2) overexpression, there is a more recently established role for the addition of HER2+ based therapy in the form of trastuzumab. However, while immune checkpoint inhibitors (ICIs) have revolutionised the treatment of other malignancies including melanoma and renal cell carcinoma over the past decade, they have only gained a foothold in GC in more recent years. This article reviews the existing evidence for ICIs in GC as a novel therapeutic option. It also looks to ongoing trials of immune checkpoint inhibition both in the perioperative and advanced setting, and in combination with other therapeutic targets including HER2+. Other investigational immune based therapies including chimeric antigen receptor T-cell (CAR-T) therapy and anti-T cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain (anti-TIGIT) therapy are considered, in addition to reviewing the building evidence for alternative therapeutic targets currently under investigation in GC, including fibroblast growth factor receptor 2b (FGFR2b) and claudin 18.2 amongst others. These novel and evolving targets represent a brave new world in therapeutic intervention in GC, with the potential to transform outcomes for patients internationally.


Subject(s)
Melanoma , Stomach Neoplasms , Humans , Stomach Neoplasms/drug therapy , Trastuzumab/therapeutic use , Melanoma/drug therapy
18.
J Immunother Cancer ; 11(2)2023 02.
Article in English | MEDLINE | ID: mdl-36754451

ABSTRACT

BACKGROUND: Oncolytic immunotherapy represents a unique therapeutic platform for the treatment of cancer. Here, we evaluated the safety and efficacy of the combination of pexastimogene devacirepvec (PexaVec) plus durvalumab (anti-programmed death ligand 1) with and without tremelimumab (anti-cytotoxic T-lymphocyte associated protein 4) in patients with standard chemotherapy refractory mismatch repair proficient (pMMR) metastatic colorectal cancer (mCRC) in a phase I/II trial. METHODS: Adult patients with histologically confirmed advanced pMMR mCRC, who had progressed on at least two prior lines of systemic chemotherapy were studied in four cohorts. Patients received four doses of PexaVec IV at a dose of 3×108 plaque forming units (pfu) (dose level 1) or 1×109 pfu (dose level 2) every 2 weeks. Twelve days after the first PexaVec administration, patients received either 1500 mg of durvalumab every 28 days alone or an additional single dose of 300 mg tremelimumab on day 1. Responses were assessed every 8 weeks by CT or MRI. AEs were recorded. The primary endpoints were safety and feasibility. Secondary endpoints included progression-free survival (PFS) and overall survival. Paired tumor samples and peripheral blood were collected to perform immune monitoring. RESULTS: Thirty-four patients with mCRC enrolled on to the study: 16 patients in the PexaVec/durvalumab cohorts and 18 patients in the PexaVec/durvalumab/tremelimumab cohorts. Overall, the combination of PexaVec plus immune checkpoint inhibitors did not result in any unexpected toxicities. Most common toxicities observed were fever and chills after PexaVec infusion. Two cases of grade 3 colitis, one case of a grade 2 myositis and one case of grade 3 hypotension resulted in discontinuation of immune checkpoint inhibitor and PexaVec treatment, respectively. The median PFS in the PexaVec/durvalumab/tremelimumab cohorts was 2.3 months (95% CI: 2.2 to 3.2 months) vs 2.1 months (95% CI: 1.7 to 2.8 months; p=0.57) in the PexaVec/durvalumab cohorts. Flow cytometry analysis of peripheral blood mononuclear cells revealed an increase in Ki67+CD8+ T cells on treatment. CONCLUSION: PexaVec in combination with durvalumab and tremelimumab is safe and tolerable. No unexpected toxicities were observed. The combination of PexaVec/durvalumab/tremelimumab demonstrated potential clinical activity in patients with pMMR mCRC, but further studies are needed to identify the predictive biomarkers. TRIAL REGISTRATION NUMBER: NCT03206073.


Subject(s)
Colorectal Neoplasms , Immune Checkpoint Inhibitors , Adult , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , CD8-Positive T-Lymphocytes , Leukocytes, Mononuclear , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/etiology
19.
Immunology ; 136(2): 176-83, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22304731

ABSTRACT

Myeloid-derived suppressor cells (MDSC) are a heterogeneous population of cells that negatively regulate the immune response during tumour progression, inflammation and infection. Only limited data are available on human MDSC because of the lack of specific markers. We have identified members of the S100 protein family-S100A8, S100A9 and S100A12 - specifically expressed in CD14(+) HLA-DR(-/low) MDSC. S100A9 staining in combination with anti-CD14 could be used to identify MDSC in whole blood from patients with colon cancer. An increase in the population of CD14(+) S100A9(high) MDSC was observed in the peripheral blood from colon cancer patients in comparison with healthy controls. Finally, nitric oxide synthase expression, a hallmark of MDSC, was induced in CD14(+) S100A9(high) upon lipopolysaccharide/interferon-γ stimulation. We propose S100 proteins as useful markers for the analysis and further characterization of human MDSC.


Subject(s)
Calgranulin B/immunology , Leukocytes, Mononuclear/immunology , Myeloid Cells/immunology , Biomarkers/blood , Calgranulin A/biosynthesis , Calgranulin A/immunology , Calgranulin B/biosynthesis , Colonic Neoplasms/blood , Colonic Neoplasms/immunology , Female , Humans , Interferon-gamma/biosynthesis , Lipopolysaccharide Receptors/analysis , Lipopolysaccharide Receptors/immunology , Lipopolysaccharides/pharmacology , Male , Middle Aged , Myeloid Cells/metabolism , Nitric Oxide Synthase/biosynthesis , S100 Proteins/biosynthesis , S100 Proteins/immunology , S100A12 Protein
20.
Int J Surg Case Rep ; 97: 107375, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35849901

ABSTRACT

INTRODUCTION: Primary and metastatic malignancies of the clavicle are extremely uncommon and difficult to treat. It carries a high morbidity rate. Total or partial claviculectomy is the recommended treatment option. PRESENTATION OF CASE: A 59-year-old male was admitted with a large left clavicular huge mass; biopsy showed poorly differentiated adenocarcinoma. Treatment started with chemoradiotherapy followed by complete excision of the tumor surgically. DISCUSSION: For treating this rare, difficult case, partial claviculectomy performed by a multispecialty team integrating three surgical skill sets: thoracic, ENT, and orthopedic surgeons, yields the best results. Clavicular carcinoma of known primary (CUP) is rare, and chemoradiotherapy showed preferable results in management. CONCLUSION: MDT reviews with surgeons, radiologists, pathologists, oncologists, and radiation oncologists are required in rare and complex cases of clavicular malignancy. Multispecialty surgical involvement is required for the safe excision of clavicular tumors with appropriate oncologic margins and fewer complications.

SELECTION OF CITATIONS
SEARCH DETAIL