Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Abdom Radiol (NY) ; 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38886219

ABSTRACT

Gastric cancer is rising in prevalence associated with high mortality, primarily due to late-stage detection, underscoring the imperative for early and precise diagnosis. Etiology involves an interplay of genetic susceptibilities and environmental factors with a prominent role of Helicobacter pylori infection. Due to its often-delayed symptom presentation, prompt and accurate diagnosis is necessary. A multimodal imaging approach, including endoscopic ultrasound (EUS), multi-detector computed tomography (MDCT), and magnetic resonance imaging (MRI) is critical for accurate staging. Each modality contributes unique advantages and limitations, highlighting the importance of integrating diagnostic strategy. Moreover, multidisciplinary conferences offer a vital collaborative platform, bringing together specialists from diverse fields for treatment planning. This synergistic approach not only enhances diagnostic precision but also improves patient outcome. This review highlights the critical role of imaging in diagnosis, staging, and management and advocates for interdisciplinary collaboration in early detection and comprehensive management of gastric cancer, aiming to reduce mortality.

2.
Cancer Res Commun ; 3(8): 1672-1677, 2023 08.
Article in English | MEDLINE | ID: mdl-37645623

ABSTRACT

Purpose: Treatment of advanced pancreatic cancer with a single therapeutic at a maximal dose has been largely ineffective at increasing survival. Combination therapies are commonly studied but often limited by toxicity. We previously showed that low-dose multiagent therapy with gemcitabine, docetaxel (taxotere), capecitabine (xeloda), and cisplatin (GTX-C) was safe, well tolerated, and effective (NCT01459614). Here, we hypothesize that adding irinotecan to GTX-C may improve survival with minimal toxicity. Experimental Design: Patients with treatment-naïve metastatic pancreatic adenocarcinoma were treated with gemcitabine, docetaxel (taxotere), capecitabine (xeloda), cisplatin, and irinotecan (GTX-CI). Treatment consisted of capecitabine 500 mg twice daily on days 1-14 and gemcitabine 300 to 500 mg/m2, docetaxel 20 mg/m2, cisplatin 15 to 20 mg/m2, and irinotecan 20 to 60 mg/m2 on days 4 and 11 of a 21-day cycle. The primary objective was 9-month overall survival (OS). Secondary objectives included response rate (RR), disease control rate (DCR), progression-free survival (PFS), and OS. Results: The regimen was well tolerated. The recommended phase II dose was gemcitabine 500 mg/m2, docetaxel 20 mg/m2, capecitabine 500 mg po bid, cisplatin 20 mg/m2, and irinotecan 20 mg/m2. Median follow-up in phase II was 11.02 months (2.37-45.17). Nine-month OS rate was 57% [95% confidence interval (CI): (41-77)]. RR was 57% [95% CI: (37-75) 50% PR and 7% CR]. DCR was 87% [95% CI: (69-96)]. Median OS and PFS were 11.02 [95% CI: (8.54-21.09)] and 8.34 [95% CI: (6.34-NA)] months, respectively. Conclusions: The addition of irinotecan to GTX-C was safe and well tolerated. While the study did not meet its primary objective, the responses were clinically meaningful using a well-tolerated regimen. Significance: We aimed to optimize the previously reported efficacious regimen of low-dose multiagent therapy with GTX-C for the treatment of metastatic pancreatic ductal adenocarcinoma by adding irinotecan. The primary objective was not met, but GTX-CI was well tolerated. The RR of 57%, median PFS of 8.3 months, median OS of 11 months, and 36-month OS rate of 19% suggest clinical benefit. Further optimization of this regimen is warranted.


Subject(s)
Adenocarcinoma , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/drug therapy , Docetaxel , Irinotecan , Capecitabine/adverse effects , Cisplatin/therapeutic use , Gemcitabine , Adenocarcinoma/drug therapy , Pancreatic Neoplasms
3.
J Immunother Cancer ; 11(6)2023 06.
Article in English | MEDLINE | ID: mdl-37286304

ABSTRACT

Gastrointestinal (GI) cancers, including esophageal, gastroesophageal junction, gastric, duodenal and distal small bowel, biliary tract, pancreatic, colon, rectal, and anal cancer, comprise a heterogeneous group of malignancies that impose a significant global burden. Immunotherapy has transformed the treatment landscape for several GI cancers, offering some patients durable responses and prolonged survival. Specifically, immune checkpoint inhibitors (ICIs) directed against programmed cell death protein 1 (PD-1), either as monotherapies or in combination regimens, have gained tissue site-specific regulatory approvals for the treatment of metastatic disease and in the resectable setting. Indications for ICIs in GI cancer, however, have differing biomarker and histology requirements depending on the anatomic site of origin. Furthermore, ICIs are associated with unique toxicity profiles compared with other systemic treatments that have long been the mainstay for GI cancer, such as chemotherapy. With the goal of improving patient care by providing guidance to the oncology community, the Society for Immunotherapy of Cancer (SITC) convened a panel of experts to develop this clinical practice guideline on immunotherapy for the treatment of GI cancer. Drawing from published data and clinical experience, the expert panel developed evidence- and consensus-based recommendations for healthcare professionals using ICIs to treat GI cancers, with topics including biomarker testing, therapy selection, and patient education and quality of life considerations, among others.


Subject(s)
Gastrointestinal Neoplasms , Quality of Life , Humans , Gastrointestinal Neoplasms/drug therapy , Immunotherapy , Societies, Medical
4.
Cancer Res Commun ; 2(11): 1418-1425, 2022 11.
Article in English | MEDLINE | ID: mdl-36970054

ABSTRACT

The treatment of metastatic pancreatic ductal adenocarcinoma (PDAC) is frequently characterized by significant toxicity and rapid development of resistance to current approved therapies. More reliable biomarkers of response are needed to guide clinical decision making. We evaluated cell-free DNA (cfDNA) using a tumor-agnostic platform and traditional biomarkers (CEA and CA19-9) levels in 12 patients treated at Johns Hopkins University on NCT02324543 "Study of Gemcitabine/Nab-Paclitaxel/Xeloda (GAX) in Combination With Cisplatin and Irinotecan in Subjects With Metastatic Pancreatic Cancer." The pretreatment values, levels after 2 months of treatment, and change in biomarker levels with treatment were compared with clinical outcomes to determine their predictive value. The variant allele frequency (VAF) of KRAS and TP53 mutations in cfDNA after 2 months of treatment was predictive of progression-free survival (PFS) and overall survival (OS). In particular, patients with a lower-than-average KRAS VAF after 2 months of treatment had a substantially longer PFS than patients with higher posttreatment KRAS VAF (20.96 vs. 4.39 months). Changes in CEA and CA19-9 after 2 months of treatment were also good predictors of PFS. Comparison via concordance index demonstrated KRAS or TP53 VAF after 2 months of treatment to be better predictors of PFS and OS than CA19-9 or CEA. This pilot study requires validation but suggests cfDNA measurement is a useful adjunct to traditional protein biomarkers and imaging evaluation and could distinguish between patients who are likely to achieve prolonged responses versus those that will have early progression and may benefit from a change in treatment approach. Significance: We report on the association of cfDNA with response durability for patients undergoing treatment with a novel metronomic chemotherapy regimen (gemcitabine, nab-paclitaxel, capecitabine, cisplatin, irinotecan; GAX-CI) for metastatic PDAC. This investigation offers encouraging evidence that cfDNA may prove to be a valuable diagnostic tool to guide clinical management.


Subject(s)
Carcinoma, Pancreatic Ductal , Cell-Free Nucleic Acids , Pancreatic Neoplasms , Humans , Irinotecan/therapeutic use , Cisplatin/therapeutic use , Deoxycytidine/therapeutic use , Cell-Free Nucleic Acids/genetics , CA-19-9 Antigen/therapeutic use , Pilot Projects , Proto-Oncogene Proteins p21(ras)/genetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Pancreatic Neoplasms/drug therapy , Carcinoma, Pancreatic Ductal/drug therapy , Capecitabine , Pancreatic Neoplasms
5.
Curr Oncol ; 30(1): 75-84, 2022 12 21.
Article in English | MEDLINE | ID: mdl-36661655

ABSTRACT

Solid organ transplants are associated with a modestly increased risk of colorectal cancers (CRC). However, the molecular profile of these cancers has not been described. We hypothesized that transplant-related immunosuppression may promote development of more immunogenic tumors as suggested by a high tumor mutation burden or mismatch repair deficiency. We performed an electronic medical record search for patients seen in the Johns Hopkins University Health System (JHHS) between 2017 and 2022 who developed CRC following solid organ transplantation. A comparator cohort of patients treated for CRC at JHHS with molecular profiling data was also identified. In this case, 29 patients were identified that developed post-transplant CRC (renal transplant, n = 18; liver transplant, n = 8; kidney-liver transplantation, n = 3). Compared to the JHHS general population CRC cohort, patients who developed post-transplant CRC had a higher rate of mismatch repair deficiency (41% versus 12%, p-value = 0.0038), and elevated tumor mutation burden (median of 22 mut/Mb versus 3.5 mut/Mb, p-value = 0.033) (range 3.52-53.65). Post-transplant tumors were enriched for PIK3CA mutations (43% versus 24%, p-value = 0.042). Post-Transplant CRCs are associated with clinical and molecular features of immune sensitivity, supporting a potential role for impaired immune surveillance in shaping the landscape of CRCs. These results may help inform the management of patients with post-transplant CRC.


Subject(s)
Colorectal Neoplasms , Liver Transplantation , Neoplastic Syndromes, Hereditary , Humans , Colorectal Neoplasms/genetics , Colorectal Neoplasms/surgery , Colorectal Neoplasms/pathology , Neoplastic Syndromes, Hereditary/genetics , Class I Phosphatidylinositol 3-Kinases/genetics
6.
Clin Epigenetics ; 13(1): 25, 2021 02 02.
Article in English | MEDLINE | ID: mdl-33531075

ABSTRACT

Epigenetic therapies may modulate the tumor microenvironment. We evaluated the safety and optimal sequence of combination DNA methyltransferase inhibitor guadecitabine with a granulocyte macrophage-colony-stimulating-factor (GM-CSF) secreting colon cancer (CRC) vaccine (GVAX) using a primary endpoint of change in CD45RO + T cells. 18 patients with advanced CRC enrolled, 11 underwent paired biopsies and were evaluable for the primary endpoint. No significant increase in CD45RO + cells was noted. Grade 3-4 toxicities were expected and manageable. Guadecitabine + GVAX was tolerable but demonstrated no significant immunologic activity in CRC. We report a novel trial design to efficiently evaluate investigational therapies with a primary pharmacodynamic endpoint.Trial registry Clinicaltrials.gov: NCT01966289. Registered 21 October, 2013.


Subject(s)
Azacitidine/analogs & derivatives , Cancer Vaccines/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , DNA (Cytosine-5-)-Methyltransferase 1/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Azacitidine/administration & dosage , Azacitidine/adverse effects , Azacitidine/pharmacology , Azacitidine/therapeutic use , Biopsy , Cancer Vaccines/administration & dosage , Cancer Vaccines/adverse effects , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Combined Modality Therapy/methods , DNA Methylation/drug effects , Epigenomics/methods , Feasibility Studies , Female , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Immunotherapy/methods , Immunotherapy, Active/methods , Leukocyte Common Antigens/drug effects , Leukocyte Common Antigens/metabolism , Male , Middle Aged , Safety , Severity of Illness Index , Tumor Microenvironment
7.
Clin Cancer Res ; 27(5): 1278-1286, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33277370

ABSTRACT

PURPOSE: Immunotherapy is currently ineffective for nearly all pancreatic ductal adenocarcinomas (PDAC), largely due to its tumor microenvironment (TME) that lacks antigen-experienced T effector cells (Teff). Vaccine-based immunotherapies are known to activate antigen-specific Teffs in the peripheral blood. To evaluate the effect of vaccine therapy on the PDAC TME, we designed a neoadjuvant and adjuvant clinical trial of an irradiated, GM-CSF-secreting, allogeneic PDAC vaccine (GVAX). PATIENTS AND METHODS: Eighty-seven eligible patients with resectable PDAC were randomly assigned (1:1:1) to receive GVAX alone or in combination with two forms of low-dose cyclophosphamide. Resected tumors following neoadjuvant immunotherapy were assessed for the formation of tertiary lymphoid aggregates (TLA) in response to treatment. The clinical endpoints are disease-free survival (DFS) and overall survival (OS). RESULTS: The neoadjuvant treatment with GVAX either alone or with two forms of low-dose cyclophosphamide is safe and feasible without adversely increasing the surgical complication rate. Patients in Arm A who received neoadjuvant and adjuvant GVAX alone had a trend toward longer median OS (35.0 months) than that (24.8 months) in the historical controls who received adjuvant GVAX alone. However, Arm C, who received low-dose oral cyclophosphamide in addition to GVAX, had a significantly shorter DFS than Arm A. When comparing patients with OS > 24 months to those with OS < 15 months, longer OS was found to be associated with higher density of intratumoral TLA. CONCLUSIONS: It is safe and feasible to use a neoadjuvant immunotherapy approach for PDACs to evaluate early biologic responses. In-depth analysis of TLAs is warranted in future neoadjuvant immunotherapy clinical trials.


Subject(s)
Adjuvants, Vaccine/administration & dosage , Cancer Vaccines/administration & dosage , Carcinoma, Pancreatic Ductal/mortality , Cyclophosphamide/administration & dosage , Lymphocytes/pathology , Neoadjuvant Therapy/mortality , Pancreatic Neoplasms/mortality , Aged , Antineoplastic Agents, Alkylating/administration & dosage , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Feasibility Studies , Female , Follow-Up Studies , Humans , Immunotherapy , Lymphocytes/drug effects , Male , Middle Aged , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Prognosis , Survival Rate
8.
Oncotarget ; 11(21): 1929-1941, 2020 May 26.
Article in English | MEDLINE | ID: mdl-32523648

ABSTRACT

PURPOSE: Metformin combined with the mTOR inhibitor rapamycin showed potential synergistic anti-tumor activity in preclinical studies in pancreatic ductal adenocarcinoma (PDA). This phase 1b study (NCT02048384) was conducted to evaluate the feasibility and activity of metformin +/- rapamycin in the maintenance setting for unselected patients with metastatic PDA (mPDA) treated with chemotherapy. MATERIALS AND METHODS: Eligible patients with stable or responding mPDA after ≥ 6 months on chemotherapy were randomized 1:1 to metformin alone (Arm A) or with rapamycin (Arm B), stratified by prior treatment with FOLFIRINOX. Fluorodeoxyglucose (FDG) PET scans and peripheral blood mononuclear cells were obtained for exploratory analyses. RESULTS: 22 subjects (11 per arm) received treatment per protocol. Median PFS/OS were 3.5 and 13.2 months respectively, with 2 year OS rate of 37%; there were no differences between arms. No responses were observed by RECIST; however, decreases in FDG avidity and/or CA19-9 were observed in several long-term survivors. Treatment related adverse events of Grade ≥ 3 occurred in 0% vs 27% of patients in Arm A vs B and were asymptomatic hematologic or electrolyte abnormalities that were not clinically significant. Improved survival was associated with low baseline neutrophil: lymphocyte ratio, baseline lack of assessable disease by PET, and greater expansion of dendritic cells following treatment. CONCLUSIONS: Metformin +/- rapamycin maintenance for mPDA was well-tolerated and several patients achieved stable disease associated with exceptionally long survival. Further prospective studies are needed to clarify the role of these agents in the maintenance setting and to enhance patient selection for such approaches.

9.
Clin Cancer Res ; 26(19): 5129-5139, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32591464

ABSTRACT

PURPOSE: This phase II study tested granulocyte-macrophage colony-stimulating factor (GM-CSF)-allogeneic pancreatic tumor cells (GVAX) and ipilimumab in metastatic pancreatic ductal adenocarcinoma (PDA) in the maintenance setting. PATIENTS AND METHODS: Patients with PDA who were treated with front-line chemotherapy consisting of 5-fluorouracil, leucovorin, irinotecan, and oxaliplatin (FOLFIRINOX) in the metastatic setting and had ongoing response or stable disease after 8-12 doses were eligible. Patients were randomized 1:1 to treatment with GVAX and ipilimumab given every 3 weeks for four doses then every 8 weeks (Arm A) or to FOLFIRINOX continuation (Arm B). The primary objective was to compare overall survival (OS) between the two arms. RESULTS: Eighty-two patients were included in the final analysis (Arm A: 40; Arm B: 42). The study was stopped for futility after interim analysis. Median OS was 9.38 months [95% confidence interval (CI), 5.0-12.2] for Arm A and 14.7 months (95% CI, 11.6-20.0) for Arm B (HR, 1.75; P = 0.019). Using immune-related response criteria, two partial responses (5.7%) were observed in Arm A and four (13.8%) in Arm B. GVAX + ipilimumab promoted T-cell differentiation into effector memory phenotypes both in the periphery and in the tumor microenvironment and increased M1 macrophages in the tumor. CONCLUSIONS: GVAX and ipilimumab maintenance therapy did not improve OS over continuation of chemotherapy and resulted in a numerically inferior survival in metastatic PDA. However, clinical responses and biological effects on immune cells were observed. Further study of novel combinations in the maintenance treatment of metastatic PDA is feasible.


Subject(s)
Adenocarcinoma/drug therapy , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Pancreatic Neoplasms/drug therapy , Tumor Microenvironment/drug effects , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cancer Vaccines/administration & dosage , Cancer Vaccines/adverse effects , Combined Modality Therapy , Disease-Free Survival , Female , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ipilimumab/administration & dosage , Ipilimumab/adverse effects , Irinotecan/administration & dosage , Irinotecan/adverse effects , Leucovorin/administration & dosage , Leucovorin/adverse effects , Macrophages/drug effects , Macrophages/immunology , Male , Middle Aged , Neoplasm Metastasis , Oxaliplatin/administration & dosage , Oxaliplatin/adverse effects , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology
10.
Am J Hematol ; 94(10): 1098-1103, 2019 10.
Article in English | MEDLINE | ID: mdl-31292986

ABSTRACT

Light chain (AL) amyloidosis is a protein folding disorder that can affect many different organ systems, in addition to the coagulation pathway. D-dimer, a measurement of fibrin degradation, is commonly elevated in hematologic malignancies, but the prevalence and significance of D-dimer elevation in AL amyloidosis is unknown. We conducted an analysis of 921 patients with AL amyloidosis that presented to the Boston University Amyloidosis Center. Baseline characteristics and laboratory data of the 897 patients included in the final cohort were analyzed. Four hundred twenty three patients (47%) had an elevated D-dimer (>0.5 µg/mL). Multivariate analysis demonstrated that a normal D-dimer level of ≤0.5 µg/mL, and a level of >0.5 µg/mL but <1 µg/mL, conferred a lower risk of mortality (HR 0.49 and 0.59, respectively) when compared to a D-dimer level ≥ 1 µg/mL. The increased risk of mortality in patients with a D-dimer level ≥ 1 µg/mL was present in all cardiac stages. The median overall survival based on D-dimer range of ≤0.5, >0.5 but <1, and ≥ 1 µg/mL was 5.86, 4.04, and 2.08 years, respectively (P < .001). This retrospective analysis demonstrates the high prevalence of D-dimer elevation in AL amyloidosis and confirms that this laboratory finding is independently associated with decreased survival.


Subject(s)
Fibrin Fibrinogen Degradation Products/analysis , Immunoglobulin Light-chain Amyloidosis/blood , Adult , Aged , Aged, 80 and over , Female , Humans , Immunoglobulin Light-chain Amyloidosis/complications , Immunoglobulin Light-chain Amyloidosis/mortality , Immunoglobulin Light-chain Amyloidosis/pathology , Kaplan-Meier Estimate , Kidney/pathology , Male , Middle Aged , Myocardium/pathology , Organ Specificity , Prevalence , Prognosis , Proportional Hazards Models , Pulmonary Embolism/etiology , Pulmonary Embolism/mortality , Retrospective Studies , Severity of Illness Index , Thrombophilia/etiology , Venous Thromboembolism/etiology , Venous Thromboembolism/mortality , Young Adult
11.
J Clin Invest ; 128(10): 4236-4242, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30272580

ABSTRACT

A complex DNA repair machinery has evolved to protect genomic integrity in the face of a myriad of DNA damage sources. When DNA repair fails, this damage can lead to carcinogenesis and tumor genomic instability. Indeed, many heritable cancer predisposition syndromes are attributable to germline defects in DNA repair pathways. On the other hand, these defects may also portend particular vulnerabilities of the cancer and may be exploited therapeutically. Most recently this has been demonstrated in the case of mismatch repair-deficient cancers, in which the immune checkpoint inhibitors have been demonstrated to be highly active. This observation has paved the way for further research investigating other sources of genomic instability that may serve as biomarkers to select patients for immunotherapy.


Subject(s)
DNA Damage , DNA Repair/immunology , Genomic Instability , Immunotherapy/methods , Neoplasms , Humans , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy
12.
Science ; 357(6349): 409-413, 2017 07 28.
Article in English | MEDLINE | ID: mdl-28596308

ABSTRACT

The genomes of cancers deficient in mismatch repair contain exceptionally high numbers of somatic mutations. In a proof-of-concept study, we previously showed that colorectal cancers with mismatch repair deficiency were sensitive to immune checkpoint blockade with antibodies to programmed death receptor-1 (PD-1). We have now expanded this study to evaluate the efficacy of PD-1 blockade in patients with advanced mismatch repair-deficient cancers across 12 different tumor types. Objective radiographic responses were observed in 53% of patients, and complete responses were achieved in 21% of patients. Responses were durable, with median progression-free survival and overall survival still not reached. Functional analysis in a responding patient demonstrated rapid in vivo expansion of neoantigen-specific T cell clones that were reactive to mutant neopeptides found in the tumor. These data support the hypothesis that the large proportion of mutant neoantigens in mismatch repair-deficient cancers make them sensitive to immune checkpoint blockade, regardless of the cancers' tissue of origin.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Biomarkers, Tumor/antagonists & inhibitors , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Colorectal Neoplasms/immunology , Colorectal Neoplasms/therapy , Neoplastic Syndromes, Hereditary/immunology , Neoplastic Syndromes, Hereditary/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Antigens, Neoplasm/immunology , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Cell Cycle Checkpoints/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , DNA Mismatch Repair , Disease-Free Survival , Female , Humans , Male , Middle Aged , Mutation , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/mortality , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes/immunology , Young Adult
13.
J Natl Compr Canc Netw ; 15(3): 401-410, 2017 03.
Article in English | MEDLINE | ID: mdl-28275038

ABSTRACT

Colorectal cancer (CRC) is a leading cause of cancer-related mortality in the United States. Response rates to second- and third-line therapy for metastatic CRC (mCRC) remain low, and immunotherapy is an attractive strategy for treatment in these patients given generally better tolerability than conventional chemotherapy and the potential for long-lasting durable responses. In particular, the novel checkpoint inhibitors (CPIs) have demonstrated unprecedented clinical activity in a wide range of cancers. The observation of clinical activity in microsatellite instability-high (MSI-H) mCRC was the first indication of a potential for CRC to respond to these agents, and has led to a breakthrough designation by the FDA for CPI use in this subset. Despite this, a proportion of MSI-H and nearly all microsatellite stable disease will not respond to single-agent checkpoint inhibition, and clinical trials are ongoing to increase responses to immunotherapy in mCRC through both better patient selection and novel combinations of immunotherapeutic agents. This review will provide a focused update on the most compelling clinical results of immunotherapy in CRC to date, as well as a summary of current strategies being tested in clinical trials in increase responses to immunotherapy in CRC.


Subject(s)
Colorectal Neoplasms/therapy , Immunotherapy , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Biomarkers, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Humans , Immunity, Innate/drug effects , Immunity, Innate/genetics , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Immunomodulation/drug effects , Immunotherapy/methods , Immunotherapy, Adoptive/methods , Microsatellite Instability , Molecular Targeted Therapy , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Treatment Outcome , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
14.
Haematologica ; 101(1): 86-90, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26452981

ABSTRACT

Patients with immunoglobulin light chain amyloidosis are at risk for both thrombotic and bleeding complications. While the hemostatic defects have been extensively studied, less is known about thrombotic complications in this disease. This retrospective study examined the frequency of venous thromboembolism in 929 patients with immunoglobulin light chain amyloidosis presenting to a single referral center, correlated risk of venous thromboembolism with clinical and laboratory factors, and examined complications of anticoagulation in this population. Sixty-five patients (7%) were documented as having at least one venous thromboembolic event. Eighty percent of these patients had events within one year prior to or following diagnosis. Lower serum albumin was associated with increased risk of VTE, with a hazard ratio of 4.30 (CI 1.60-11.55; P=0.0038) for serum albumin less than 3 g/dL compared to serum albumin greater than 4 g/dL. Severe bleeding complications were observed in 5 out of 57 patients with venous thromboembolism undergoing treatment with anticoagulation. Prospective investigation should be undertaken to better risk stratify these patients and to determine the optimal strategies for prophylaxis against and management of venous thromboembolism.


Subject(s)
Amyloidosis , Hemorrhage , Immunoglobulin Light Chains/blood , Serum Albumin/metabolism , Venous Thromboembolism , Adult , Aged , Aged, 80 and over , Amyloidosis/blood , Amyloidosis/complications , Female , Hemorrhage/blood , Hemorrhage/etiology , Humans , Male , Middle Aged , Risk Factors , Venous Thromboembolism/blood , Venous Thromboembolism/etiology
15.
HPB (Oxford) ; 17(4): 292-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25250696

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma (PDA) is comprised of a prominent desmoplastic stromal compartment and only 10-40% of the tumour consists of PDA cells. However, how stromal components should be assessed and how the characteristics of the stromal compartment determine clinical outcomes in PDA patients remain unknown. METHOD: A cohort of 66 consecutive patients who underwent pancreaticoduodenectomy and were primarily followed at Johns Hopkins Hospital between 1998 and 2004, and treated with adjuvant therapy, were included in a retrospective analysis. Resected PDA blocks with good tissue preservation were available for all patients. A new, computer-aided, quantitative method was developed to assess the density and activity of stroma in PDAs and the associations of these characteristics with clinical outcomes. RESULTS: High stromal density in resected PDA was found to be significantly associated with longer disease-free [adjusted hazard ratio (aHR) 0.39; P = 0.001] and overall (aHR 0.44; P = 0.004) survival after adjusting for the use of pancreatic cancer vaccine therapy, as well as gender and resection margin positivity. Stromal activity, representing activated pancreatic stellate cells in PDAs, was not significantly associated with the prognosis of resected PDAs. CONCLUSIONS: These results illustrate the complexity of the role of stroma in PDAs. Further exploration of the prognostic ability of the characteristics of stroma is warranted.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/therapy , Chemoradiotherapy, Adjuvant , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , Pancreaticoduodenectomy , Stromal Cells/pathology , Actins/analysis , Adult , Aged , Aged, 80 and over , Baltimore , Biomarkers, Tumor/analysis , Carcinoma, Pancreatic Ductal/chemistry , Carcinoma, Pancreatic Ductal/mortality , Collagen/analysis , Disease-Free Survival , Female , Humans , Image Interpretation, Computer-Assisted , Immunohistochemistry , Kaplan-Meier Estimate , Keratins/analysis , Male , Middle Aged , Neoplasm, Residual , Pancreatic Neoplasms/chemistry , Pancreatic Neoplasms/mortality , Predictive Value of Tests , Proportional Hazards Models , Retrospective Studies , Risk Factors , Stromal Cells/chemistry , Time Factors , Treatment Outcome
16.
J Vis Exp ; (71)2013 Jan 03.
Article in English | MEDLINE | ID: mdl-23328703

ABSTRACT

B7-H1/PD-L1, a member of the B7 family of immune-regulatory cell-surface proteins, plays an important role in the negative regulation of cell-mediated immune responses through its interaction with its receptor, programmed death-1 (PD-1). Overexpression of B7-H1 by tumor cells has been noted in a number of human cancers, including melanoma, glioblastoma, and carcinomas of the lung, breast, colon, ovary, and renal cells, and has been shown to impair anti-tumor T-cell immunity. Recently, B7-H1 expression by pancreatic adenocarcinoma tissues has been identified as a potential prognostic marker. Additionally, blockade of B7-H1 in a mouse model of pancreatic cancer has been shown to produce an anti-tumor response. These data suggest the importance of B7-H1 as a potential therapeutic target. Anti-B7-H1 blockade antibodies are therefore being tested in clinical trials for multiple human solid tumors including melanoma and cancers of lung, colon, kidney, stomach and pancreas. In order to eventually be able to identify the patients who will benefit from B7-H1 targeting therapies, it is critical to investigate the correlation between expression and localization of B7-H1 and patient response to treatment with B7-H1 blockade antibodies. Examining the expression of B7-H1 in human pancreatic adenocarcinoma tissues through immunohistochemistry will give a better understanding of how this co-inhibitory signaling molecule contributes to the suppression of antitumor immunity in the tumor's microenvironment. The anti-B7-H1 monoclonal antibody (clone 5H1) developed by Chen and coworkers has been shown to produce reliable staining results in cryosections of multiple types of human neoplastic tissues, but staining on paraffin-embedded slides had been a challenge until recently. We have developed the B7-H1 staining protocol for paraffin-embedded slides of pancreatic adenocarcinoma tissues. The B7-H1 staining protocol described here produces consistent membranous and cytoplasmic staining of B7-H1 with little background.


Subject(s)
Adenocarcinoma/chemistry , B7-H1 Antigen/analysis , Immunohistochemistry/methods , Pancreatic Neoplasms/chemistry , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , B7-H1 Antigen/metabolism , Humans , Mice , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Paraffin Embedding
17.
Biochemistry ; 45(24): 7511-21, 2006 Jun 20.
Article in English | MEDLINE | ID: mdl-16768447

ABSTRACT

Sirtuins comprise a family of enzymes that catalyze the deacetylation of acetyllysine side chains in a reaction that consumes NAD+. Although several crystal structures of sirtuins bound to non-native acetyl peptides have been determined, relatively little about how sirtuins discriminate among different substrates is understood. We have carried out a systematic structural and thermodynamic analysis of several peptides bound to a single sirtuin, the Sir2 homologue from Thermatoga maritima (Sir2Tm). We report structures of five different forms of Sir2Tm: two forms bound to the p53 C-terminal tail in the acetylated and unacetylated states, two forms bound to putative acetyl peptide substrates derived from the structured domains of histones H3 and H4, and one form bound to polypropylene glycol (PPG), which resembles the apoenzyme. The structures reveal previously unobserved complementary side chain interactions between Sir2Tm and the first residue N-terminal to the acetyllysine (position -1) and the second residue C-terminal to the acetyllysine (position +2). Isothermal titration calorimetry was used to compare binding constants between wild-type and mutant forms of Sir2Tm and between additional acetyl peptide substrates with substitutions at positions -1 and +2. The results are consistent with a model in which peptide positions -1 and +2 play a significant role in sirtuin substrate binding. This model provides a framework for identifying sirtuin substrates.


Subject(s)
Sirtuins/chemistry , Sirtuins/metabolism , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Escherichia coli/genetics , Models, Molecular , Molecular Sequence Data , Peptides/chemistry , Peptides/metabolism , Protein Binding , Sequence Homology, Amino Acid , Sirtuins/genetics , Structure-Activity Relationship , Substrate Specificity , Thermodynamics , Thermotoga maritima/enzymology , Thermotoga maritima/isolation & purification
18.
Mol Cell ; 17(6): 855-68, 2005 Mar 18.
Article in English | MEDLINE | ID: mdl-15780941

ABSTRACT

Sir2 enzymes form a unique class of NAD(+)-dependent deacetylases required for diverse biological processes, including transcriptional silencing, regulation of apoptosis, fat mobilization, and lifespan regulation. Sir2 activity is regulated by nicotinamide, a noncompetitive inhibitor that promotes a base-exchange reaction at the expense of deacetylation. To elucidate the mechanism of nicotinamide inhibition, we determined ternary complex structures of Sir2 enzymes containing nicotinamide. The structures show that free nicotinamide binds in a conserved pocket that participates in NAD(+) binding and catalysis. Based on our structures, we engineered a mutant that deacetylates peptides by using nicotinic acid adenine dinucleotide (NAAD) as a cosubstrate and is inhibited by nicotinic acid. The characteristics of the altered specificity enzyme establish that Sir2 enzymes contain a single site that participates in catalysis and nicotinamide regulation and provides additional insights into the Sir2 catalytic mechanism.


Subject(s)
Gene Expression Regulation, Fungal , Histone Deacetylases/metabolism , NAD/analogs & derivatives , NAD/metabolism , Niacinamide/metabolism , Silent Information Regulator Proteins, Saccharomyces cerevisiae/metabolism , Sirtuins/chemistry , Sirtuins/metabolism , Acetylation , Amino Acid Sequence , Catalysis , Histone Deacetylases/genetics , Molecular Sequence Data , Niacin/pharmacology , Sequence Homology, Amino Acid , Silent Information Regulator Proteins, Saccharomyces cerevisiae/genetics , Sirtuin 2 , Sirtuins/genetics , Substrate Specificity , Vasodilator Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...