Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
J Cutan Pathol ; 49(9): 787-790, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35734844

ABSTRACT

Merkel cell carcinoma (MCC) is an aggressive, highly metastatic, cutaneous neuroendocrine malignancy with poor prognosis. Here, we describe a MCC excision specimen with a rare case of tumor-associated amyloid deposition in the absence of residual tumor cells. A 72-year-old man presented with a lesion of 5-6 months' duration on his left elbow, clinically thought to be a ganglion cyst. The biopsy specimen revealed a Stage IIA MCC with classic histomorphologic and immunophenotypic findings, with tumor extending to the tissue edges. The patient underwent wide local excision with negative margins and a negative sentinel lymph node biopsy. Although the patient did not receive any presurgical chemotherapy, immunotherapy, or targeted therapy, the re-excision specimen showed only amphophilic, feathery deposits that were salmon-pink with Congo red stain and further confirmed as amyloid by electron microscopy; there were no residual carcinoma cells. Amyloid deposition in MCC has been described in rare case reports. Our case was extraordinary in that there was only amyloid deposition and an associated granulomatous reaction, without identifiable MCC cells. This case demonstrates that amyloid deposition may be evidence of a prior MCC at the site of a prior procedure and may warrant careful evaluation for residual MCC.


Subject(s)
Carcinoma, Merkel Cell , Skin Neoplasms , Aged , Carcinoma, Merkel Cell/pathology , Humans , Male , Sentinel Lymph Node Biopsy , Skin/pathology , Skin Neoplasms/pathology
2.
Clin Cancer Res ; 26(8): 1886-1895, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32015020

ABSTRACT

PURPOSE: There is a need for sensitive, reproducible biomarkers for patients with stage III melanoma to guide clinical decision making. Circulating tumor cells (CTCs) can be detected in patients with melanoma; however, there are limited data regarding their significance in stage III disease. The aim of this study was to determine whether CTCs are associated with early relapse in stage III melanoma. EXPERIMENTAL DESIGN: We prospectively assessed CTCs at first presentation in clinic (baseline) for 243 patients with stage III melanoma. CTCs were measured using the CellSearch System. Relapse-free survival (RFS) was compared between patients with one or more baseline CTC versus those with no CTCs. Log-rank test and Cox regression analysis were applied to establish associations of CTCs with RFS. RESULTS: At least one baseline CTC was identified in 90 of 243 (37%) patients. Forty-five (19%), 67 (28%), 118 (49%), and 13 (5%) patients were stage IIIA, IIIB, IIIC, or IIID, respectively. CTC detection was not associated with substage, or primary tumor characteristics. Multivariable analysis demonstrated that the detection of ≥1 baseline CTC was significantly associated with decreased 6-month RFS [log-rank, P < 0.0001; HR, 3.62, 95% confidence interval (CI), 1.78-7.36; P < 0.0001] and 54-month RFS (log-rank, P = 0.01; HR, 1.69; 95% CI, 1.13-2.54; P = 0.01). CONCLUSIONS: ≥1 CTC was independently associated with melanoma relapse, suggesting that CTC assessment may be useful to identify patients at risk for relapse who could derive benefit from adjuvant therapy.


Subject(s)
Biomarkers, Tumor/blood , Lymph Nodes/pathology , Melanoma/pathology , Neoplasm Recurrence, Local/pathology , Neoplastic Cells, Circulating/pathology , Skin Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Disease Progression , Female , Humans , Lymphatic Metastasis , Male , Melanoma/blood , Middle Aged , Neoplasm Recurrence, Local/blood , Neoplasm Staging , Prospective Studies , Skin Neoplasms/blood , Survival Rate , Melanoma, Cutaneous Malignant
3.
J Clin Oncol ; 38(13): 1429-1441, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31990608

ABSTRACT

PURPOSE: Improved understanding of the incidence, risk factors, and timing of CNS metastasis is needed to inform surveillance strategies for patients with melanoma. PATIENTS AND METHODS: Clinical data were extracted from the databases of 2 major melanoma centers in the United States and Australia for 1,918 patients with American Joint Committee on Cancer (AJCC) 8th edition stage III melanoma, diagnosed from 1998-2014, who had (negative) baseline CNS imaging within 4 months of diagnosis. The cumulative incidence of CNS metastasis was calculated in the presence of the competing risk of death, from stage III presentation and at benchmark time points 1, 2, and 5 years postdiagnosis. RESULTS: At a median follow-up of 70.2 months, distant recurrence occurred in 711 patients (37.1%). The first site of distant metastasis was CNS only for 3.9% of patients, CNS and extracranial (EC) for 1.8%, and EC only for 31.4%. Overall, 16.7% of patients were diagnosed with CNS metastasis during follow-up. The cumulative incidence of CNS metastasis was 3.6% (95% CI, 2.9% to 4.6%) at 1 year, 9.6% (95% CI, 8.3% to 11.0%) at 2 years, and 15.8% (95% CI, 14.1% to 17.6%) at 5 years. The risk of CNS metastasis was significantly influenced by patient sex, age, AJCC stage, primary tumor site, and primary tumor mitotic rate in multivariable and conditional analyses. High primary tumor mitotic rate was significantly associated with increased risk of CNS metastasis at diagnosis and all subsequent time points examined. CONCLUSION: Similar rates of CNS metastasis were observed in 2 large, geographically distinct cohorts of patients with stage III melanoma. The results highlight the importance of primary tumor mitotic rate. Furthermore, they provide a framework for developing evidence-based surveillance strategies and evaluating the impact of contemporary adjuvant therapies on the risk of CNS metastasis development.


Subject(s)
Central Nervous System Neoplasms/diagnosis , Melanoma/prevention & control , Skin Neoplasms/prevention & control , Adolescent , Adult , Aged , Aged, 80 and over , Australia/epidemiology , Central Nervous System Neoplasms/epidemiology , Central Nervous System Neoplasms/secondary , Cohort Studies , Female , Humans , Incidence , Male , Melanoma/pathology , Middle Aged , Multivariate Analysis , Neoplasm Recurrence, Local , Neoplasm Staging , Prognosis , Risk Assessment/methods , Risk Assessment/statistics & numerical data , Risk Factors , Skin Neoplasms/pathology , United States/epidemiology , Young Adult
4.
Nat Rev Clin Oncol ; 15(12): 763-776, 2018 12.
Article in English | MEDLINE | ID: mdl-30287935

ABSTRACT

Merkel cell carcinoma (MCC) is a rare and aggressive skin cancer associated with advanced age and immunosuppression. Over the past decade, an association has been discovered between MCC and either integration of the Merkel cell polyomavirus, which likely drives tumorigenesis, or somatic mutations owing to ultraviolet-induced DNA damage. Both virus-positive and virus-negative MCCs are immunogenic, and inhibition of the programmed cell death protein 1 (PD-1)-programmed cell death 1 ligand 1 (PD-L1) immune checkpoint has proved to be highly effective in treating patients with metastatic MCC; however, not all patients have a durable response to immunotherapy. Despite these rapid advances in the understanding and management of patients with MCC, many basic, translational and clinical research questions remain unanswered. In March 2018, an International Workshop on Merkel Cell Carcinoma Research was held at the US National Cancer Institute, at which academic, government and industry experts met to identify the highest-priority research questions. Here, we review the biology and treatment of MCC and report the consensus-based recommendations agreed upon during the workshop.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Merkel Cell/drug therapy , Carcinoma, Merkel Cell/genetics , Immunotherapy/trends , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Carcinoma, Merkel Cell/immunology , Carcinoma, Merkel Cell/virology , Humans , Polyomavirus/genetics , Polyomavirus/pathogenicity , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology
5.
J Immunother Cancer ; 5(1): 102, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29254506

ABSTRACT

BACKGROUND: Immune related adverse events (irAEs) are associated with immunotherapy for cancer and while results suggest improvement in tumor control and overall survival in those experiencing irAEs, the long-term impact is debated. We evaluated irAE reports related to high dose interleukin-2 therapy (IL-2) documented in the PROCLAIMSM registry data base from 2008 to 2016 (NCT01415167, August 9, 2011). METHODS: Reports on 1535 patients, including 623 with metastatic melanoma (mM) and 919 with metastatic renal cell cancer (mRCC) (7 patients had both diseases), were queried for irAEs. The timing of the event was categorized as occurring before, during or after IL-2 or related to any checkpoint inhibitor (CPI). mM patients and mRCC patients were analyzed separately. Tumor control [complete + partial response + stable disease (CR + PR + SD) was compared between those experiencing no irAE versus those with the development of irAEs. Survival was analyzed by tumor type related to timing of irAE and IL-2, and in those with or without exposure to CPI. RESULTS: Median follow-up was 3.5+ years (range 1-8+ years), 152 irAEs were reported in 130 patients (8.4% of all PROCLAIMSM patients): 99 (16%) in mM and 53 (5.8%) in mRCC patients. 31 irAEs occurred prior to IL-2, 24 during IL-2, and 97 after IL-2 therapy. 74 irAEs were attributed to IL-2 only (during/ after IL-2). Of the 97 post IL-2 irAEs, 24 were attributed to CPI, and 15 could not be distinguished as caused by IL-2 or CPI. Tumor control was 71% for those experiencing irAE, and 56% for those with no irAE (p = 0.0008). Overall survival was significantly greater for those experiencing irAEs during/ after IL-2 therapy, compared to those with no irAE or irAE before IL-2 therapy, in mM patients, median 48 months vs 18 months (p < 0.0001), and in mRCC patients, median 60 months vs 40 months (p = 0.0302), independent of CPI-related irAEs. IL-2-related irAEs were primarily vitiligo and thyroid dysfunction (70% of IL-2 related irAEs), with limited further impact. CONCLUSIONS: irAEs following IL-2 therapy are associated with improved tumor control and overall survival. IrAEs resulting from IL-2 and from CPIs are qualitatively different, and likely reflect different mechanisms of action of immune activation and response.


Subject(s)
Antineoplastic Agents, Immunological/adverse effects , Carcinoma, Renal Cell/drug therapy , Interleukin-2/adverse effects , Kidney Neoplasms/drug therapy , Melanoma/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Immunological/therapeutic use , Female , Humans , Interleukin-2/therapeutic use , Male , Middle Aged , Neoplasm Metastasis , Registries , Survival Analysis , Treatment Outcome , Young Adult
6.
Clin Genitourin Cancer ; 15(1): 31-41.e4, 2017 02.
Article in English | MEDLINE | ID: mdl-27916626

ABSTRACT

BACKGROUND: This analysis describes the outcome for patients who received targeted therapy (TT) prior to or following high-dose interleukin-2 (HD IL-2). PATIENTS AND METHODS: Patients with renal cell carcinoma (n = 352) receiving HD IL-2 were enrolled in ProleukinR Observational Study to Evaluate the Treatment Patterns and Clinical Response in Malignancy (PROCLAIMSM) beginning in 2011. Statistical analyses were performed using datasets as of September 24, 2015. RESULTS: Overall, there were 4% complete response (CR), 13% partial response (PR), 39% stable disease (SD), and 43% progressive disease (PD) with HD IL-2. The median overall survival (mOS) was not reached in patients with CR, PR, or SD, and was 15.5 months in patients with PD (median follow-up, 21 months). Sixty-one patients had prior TT before HD IL-2 with an overall response rate (ORR) to HD IL-2 of 19% (1 CR, 9 PR) and an mOS of 22.1 months. One hundred forty-nine patients received TT only after HD IL-2 with an mOS of 35.5 months. One hundred forty-two patients had no TT before or after HD IL-2, and mOS was not reached. The mOS was 8.5 months in PD patients who received HD IL-2 without follow-on TT and 29.7 months in PD patients who received follow-on TT after HD IL-2. CONCLUSIONS: HD IL-2 as sole front-line therapy, in the absence of added TT, shows extended clinical benefit (CR, PR, and SD). Patients with PD after HD IL-2 appear to benefit from follow-on TT. Patients who progressed on TT and received follow-on HD IL-2 experienced major clinical benefit. HD IL-2 therapy should be considered in eligible patients.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Renal Cell/drug therapy , Interleukin-2/analogs & derivatives , Kidney Neoplasms/drug therapy , Molecular Targeted Therapy/methods , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/immunology , Disease-Free Survival , Female , Humans , Immunotherapy , Interleukin-2/administration & dosage , Interleukin-2/therapeutic use , Kidney Neoplasms/immunology , Male , Middle Aged , Neoplasm Metastasis , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use , Survival Analysis , Treatment Outcome
7.
Cancer Immunol Immunother ; 65(12): 1533-1544, 2016 12.
Article in English | MEDLINE | ID: mdl-27714434

ABSTRACT

High-dose interleukin-2 (HD IL-2) was approved for treatment of metastatic renal cell carcinoma (mRCC) in 1992 and for metastatic melanoma (mM) in 1998, in an era predating targeted therapies and immune checkpoint inhibitors. The PROCLAIMSM registry was established to collect and analyze data for patients treated with HD IL-2 in the current era. This analysis includes 170 patients with mM and 192 patients with mRCC treated between 2005 and 2012 with survival data current as of July 27, 2015. For patients with mM, complete response (CR) was observed in 5 %, partial response (PR) in 10 %, stable disease (SD) in 22 %, and 63 % had progressive disease (PD). The median overall survival (mOS) for these patients was 19.6 months, with a median follow-up of 43.1 months. The mOS was not reached for patients achieving CR or PR, and was 33.4 months for patients with SD. For patients with mRCC, 6 % achieved CR, 9 % had PR, 22 % had SD, and 62 % had PD. The mOS was 41 months, with a median follow-up of 46.6 months. The mOS for patients who had CR and PR was not reached and was 49.6 months for patients with SD. There were no treatment-related deaths among 362 patients. The duration of mOS for patients with mM and mRCC is longer than historically reported. These data support a continued role for IL-2 in the treatment of eligible patients with mM or mRCC and warrant further evaluation of HD IL-2 in combination or sequence with other therapeutic agents.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/drug therapy , Immunotherapy/methods , Interleukin-2/therapeutic use , Melanoma/drug therapy , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/pathology , Female , Humans , Interleukin-2/administration & dosage , Interleukin-2/pharmacology , Male , Melanoma/mortality , Melanoma/pathology , Middle Aged , Survival Rate , Young Adult
8.
Clin Cancer Res ; 21(3): 561-8, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25424850

ABSTRACT

PURPOSE: High-dose aldesleukin (HD IL2) received FDA approval for the treatment of metastatic renal cell carcinoma (MRCC) in 1992, producing a 14% objective response rate (ORR) and durable remissions. Retrospective studies suggested that clinical and pathologic features could predict for benefit. The Cytokine Working Group conducted this prospective trial to validate proposed predictive markers of response to HD IL2. EXPERIMENTAL DESIGN: Standard HD IL2 was administered to prospectively evaluate whether the ORR of patients with mRCC with "good" predictive pathologic features based on an "integrated selection" model [ISM (e.g., clear-cell histology subclassification and carbonic anhydrase-9 (CA-9) IHC staining] was significantly higher than the ORR of a historical, unselected population. Archived tumor was collected for pathologic analysis including tumor programmed death-ligand 1 (PD-L1) expression. RESULTS: One hundred and twenty eligible patients were enrolled between June 11 and September 7; 70% were Memorial Sloan Kettering Cancer Center (New York, NY) intermediate risk, 96% had clear cell RCC, and 99% had prior nephrectomy. The independently assessed ORR was 25% (30/120, 95% CI, 17.5%-33.7%, P = 0.0014; 3 complete responses, 27 partial responses) and was higher than a historical ORR. Thirteen patients (11%) remained progression free at 3 years and the median overall survival was 42.8 months. ORR was not statistically different by ISM classification ("good-risk" 23% vs. "poor-risk" 30%; P = 0.39). ORR was positively associated with tumor PD-L1 expression (P = 0.01) by IHC. CONCLUSIONS: In this prospective, biomarker validation study, HD IL2 produced durable remissions and prolonged survival in both "good" and "poor-risk" patients. The proposed ISM was unable to improve the selection criteria. Novel markers (e.g., tumor PD L1 expression) appeared useful, but require independent validation.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/pathology , Interleukin-2/analogs & derivatives , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Adult , Aged , Carcinoma, Renal Cell/mortality , Humans , Interleukin-2/administration & dosage , Kidney Neoplasms/mortality , Middle Aged , Neoplasm Metastasis , Prognosis , Recombinant Proteins/administration & dosage , Risk Factors , Treatment Outcome
9.
Thromb Haemost ; 113(2): 329-37, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25354586

ABSTRACT

Carbon monoxide (CO) is a gaseous autacoid known to positively regulate vascular tone; however, its role in angiogenesis is unknown. The aim of this study was to investigate the effect of CO on angiogenesis and vascular endothelial growth factor (VEGF) receptor-2 phosphorylation. Human umbilical vein endothelial cells (HUVECs) were cultured on growth factor-reduced Matrigel and treated with a CO-releasing molecule (CORM-2) or exposed to CO gas (250 ppm). Here, we report the surprising finding that exposure to CO inhibits vascular endothelial growth factor (VEGF)-induced endothelial cell actin reorganisation, cell proliferation, migration and capillary-like tube formation. Similarly, CO suppressed VEGF-mediated phosphorylation of VEGFR-2 at tyrosine residue 1175 and 1214 and basic fibroblast growth factor- (FGF-2) and VEGF-mediated Akt phosphorylation. Consistent with these data, mice exposed to 250 ppm CO (1h/day for 14 days) exhibited a marked decrease in FGF-2-induced Matrigel plug angiogenesis (p<0.05). These data establish a new biological function for CO in angiogenesis and point to a potential therapeutic use for CO as an anti-angiogenic agent in tumour suppression.


Subject(s)
Carbon Monoxide/chemistry , Neovascularization, Pathologic , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cell Movement , Cell Proliferation , Collagen/chemistry , Drug Combinations , Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells , Humans , Immunoprecipitation , Laminin/chemistry , Mice, Inbred C57BL , Organometallic Compounds/chemistry , Phosphorylation , Proteoglycans/chemistry , Recombinant Proteins/chemistry , Tyrosine/chemistry
10.
J Immunother ; 37(7): 360-5, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25075565

ABSTRACT

Although tyrosine kinase inhibitors (TKI) are the most common first-line therapy for metastatic renal cell carcinoma, high-dose interleukin-2 (HD-IL2) remains the only agent that provides durable complete responses. The optimal sequence of these agents remains uncertain. This retrospective multi-institutional study examined the safety and efficacy of HD-IL2 following TKI therapy. After IRB approval at 7 HD-IL2 centers, data relating to patient, disease, and treatment characteristics among 40 consecutive patients with metastatic renal cell carcinoma who were treated with HD-IL2 after at least 1 prior TKI therapy were retrospectively collected. The most common cardiac adverse events were grade 3 hypotension and vascular leak syndrome. Six patients (15%) experienced other grade ≥3 cardiac adverse events. There were 2 treatment-related deaths due to congestive heart failure, occurring in 1 patient with short TKI to HD-IL2 interval and another patient with an abnormal baseline cardiac stress test. Best responses included 2 CRs (5%, duration 40+ and 62+ mo), 3 PRs (8%, duration 6, 11, and 24 mo), 13 SD (32%, median duration 12 mo), 20 PD (50%), and 2 not evaluable patients. Median overall survival was 22 months. Administration of HD-IL2 could be safe and effective after TKI therapy; however, careful selection of patients is critical. We recommend baseline cardiac risk factor assessment, screening with both cardiac stress test and echocardiogram, and allowing a TKI to HD-IL2 interval of at least 2 months.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/pathology , Interleukin-2/administration & dosage , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Protein Kinase Inhibitors/therapeutic use , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Carcinoma, Renal Cell/mortality , Cardiovascular Diseases/etiology , Female , Humans , Interleukin-2/adverse effects , Kidney Neoplasms/mortality , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Prognosis , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Retrospective Studies , Risk Factors , Treatment Outcome
11.
Pharmacotherapy ; 31(1): 62-75, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21182360

ABSTRACT

Cancer therapeutics is undergoing a revolution with the advent of new drugs that can selectively target molecules responsible for carcinogenesis and tumor growth. The type and mechanism of these targeting drugs vary. Some are small molecules that specifically target a binding site on a receptor or signal transduction molecule. Antibodies have been engineered to bind to the receptors or the corresponding ligands that mediate a critical cancer activity. In almost all cases, the intent is to inhibit or shut down a specific molecular pathway. Unprecedented activity against the cancer is seen without overt traditional toxicities such as alopecia, nausea and/or vomiting, and cytopenias. Unfortunately, an increase in toxicity has now become evident as more experience accumulates with the use of these drugs. In some cases, unexpected cardiotoxicities have arisen when these new drugs have been added to more conventional chemotherapies. Heart failure is the unfortunate manifestation for many of these toxicities. We outline the scope of this problem and examine the mechanisms of drug-induced heart failure. The distinctive signs and symptoms specific to each drug are described, and the diagnosis and treatment of the condition are discussed. Our aim is to allow the practitioner to recognize the unusual manifestations of heart failure in this setting in order to make a timely diagnosis and begin appropriate treatment measures.


Subject(s)
Antibodies, Monoclonal/adverse effects , Antineoplastic Agents/adverse effects , Heart Failure/chemically induced , Molecular Targeted Therapy/adverse effects , Neoplasms/drug therapy , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Female , Heart Failure/drug therapy , Humans , Male , Neoplasms/metabolism , Prognosis , Signal Transduction/drug effects
12.
Carcinogenesis ; 31(2): 281-6, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19955395

ABSTRACT

Bladder cancer is one of the common human cancers and also has a very high recurrence rate. There is a great need for agents capable of inhibiting bladder cancer development and recurrence. Here, we report that allyl isothiocyanate (AITC), an ingredient of many common cruciferous vegetables, potently inhibited the proliferation of bladder carcinoma cell lines in vitro [half maximal inhibitory concentration (IC(50)) of 2.7-3.3 microM], which was associated with profound G(2)/M arrest and apoptosis. In contrast, AITC was markedly less toxic to normal human bladder epithelial cells (IC(50) of 69.4 microM). AITC was then evaluated in two rat bladder cancer models in vivo (an orthotopic model and a subcutaneous model). The orthotopic model closely mimics human bladder cancer development and recurrence. We show that a low oral dose of AITC (1 mg/kg) significantly inhibited the development and muscle invasion of the orthotopic bladder cancers but was ineffective against the subcutaneous xenografts of the same cancer cells in the same animals. This differential effect was explained by our finding that urinary levels of AITC equivalent were two to three orders of magnitude higher than that in the plasma and that its levels in the orthotopic cancer tissues were also three orders of magnitude higher than that in the subcutaneous cancer tissues. Moreover, we show that AITC is a multi-targeted agent against bladder cancer. In conclusion, AITC is selectively delivered to bladder cancer tissue through urinary excretion and potently inhibits bladder cancer development and invasion.


Subject(s)
Food Preservatives/pharmacology , Isothiocyanates/pharmacology , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/pathology , Animals , Apoptosis/drug effects , Blotting, Western , Cell Proliferation/drug effects , Cells, Cultured , Female , Flow Cytometry , Food Preservatives/pharmacokinetics , Humans , Isothiocyanates/pharmacokinetics , Male , Rats , Rats, Inbred F344 , Tissue Distribution , Urinary Bladder/drug effects , Urinary Bladder/pathology , Xenograft Model Antitumor Assays
14.
Pharmacotherapy ; 29(4): 473-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19323623

ABSTRACT

Cardiotoxicity is an emerging concern with a new class of drugs known as targeted agents, which include trastuzumab and sunitinib. Sunitinib is a small molecule that inhibits multiple tyrosine kinase receptors. This drug was approved by the United States Food and Drug Administration in 2006 for the treatment of clear cell metastatic renal cell carcinoma and advanced gastrointestinal stromal tumors. We describe a 65-year-old woman who was treated with sunitinib for metastatic clear cell renal cell carcinoma. After 5 months of therapy, she developed acute heart failure requiring hospitalization; sunitinib was immediately discontinued. The patient had classic symptoms of heart failure, including pleural effusion. An echocardiogram revealed a left ventricular ejection fraction of 30%. She received standard treatment for heart failure, including a beta-blocker, an angiotensin-converting enzyme inhibitor, and diuretics. Within 1 month, the patient's symptoms resolved, and subsequent cardiac evaluation showed that her left ventricular ejection fraction returned to normal. According to the Common Terminology Criteria for Adverse Events developed by the National Cancer Institute, her cardiac event associated with sunitinib was defined as grade III toxicity. One month later, sorafenib, another tyrosine kinase inhibitor, was started with the aim of continuing her previous response to sunitinib. After 7 months of sorafenib therapy, the patient had no evidence of heart failure, and her condition was responding to treatment. Clinicians should be aware that sunitinib-induced heart failure occurs occultly and that many--but not all--cases resolve with discontinuation of the drug. Use of sorafenib after sunitinib-induced heart failure appears to be safe and effective, which suggests that cardiotoxicity is not a general class effect of the tyrosine kinase inhibitors.


Subject(s)
Antineoplastic Agents/adverse effects , Benzenesulfonates/therapeutic use , Carcinoma, Renal Cell/complications , Heart Failure/chemically induced , Heart Failure/drug therapy , Indoles/adverse effects , Kidney Neoplasms/complications , Protein Kinase Inhibitors/therapeutic use , Pyridines/therapeutic use , Pyrroles/adverse effects , Aged , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/secondary , Case-Control Studies , Female , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Niacinamide/analogs & derivatives , Phenylurea Compounds , Sorafenib , Sunitinib
15.
J Urol ; 181(1): 336-42, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19010488

ABSTRACT

PURPOSE: The potent effects of PAI-1 on tumorigenesis and angiogenesis in various experimental models are complex, complicated and at times contradictory. We determined the therapeutic potential of PAI-1 for inhibiting bladder tumor invasion under conditions that closely mimic the clinical setting. MATERIALS AND METHODS: An orthotopic rat bladder tumor model was established by implanting AY-27 rat transitional carcinoma cells into the bladder lumen of syngeneic Fischer F344 rats. Three weeks after implantation 1 microM PAI-1 was administrated directly into the bladder lumen twice weekly for 2 weeks. Two days after the final treatment tumor size, total bladder weight, tumor stage and angiogenesis were assessed. To assess the uPA axis the levels of active and total uPA, and active and total PAI-1 in tumor extracts were determined 0, 2, 24 and 48 hours after intravesical PAI-1 administration. RESULTS: Intravesical PAI-1 bound and inactivated its molecular target, tumor uPA. There was significant inhibition of bladder tumor progression, as manifested by 53%, 37% and 57% reductions in tumor size, total bladder weight and angiogenesis, respectively. Only 22% of PAI-1 treated tumors invaded muscle vs 79% in controls. No PAI-1 toxicity was detected. CONCLUSIONS: To our knowledge this study is the first to demonstrate that intravesical treatment with PAI-1 significantly inhibits tumor progression in an in vivo model of bladder cancer. Further clinical development is warranted for using PAI-1 directly or in combination with current standards, such as bacillus Calmette-Guerin or interferon.


Subject(s)
Plasminogen Activator Inhibitor 1/administration & dosage , Serine Proteinase Inhibitors/administration & dosage , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/prevention & control , Administration, Intravesical , Animals , Disease Progression , Neoplasm Invasiveness , Rats , Rats, Inbred F344
16.
Curr Oncol Rep ; 10(3): 259-63, 2008 May.
Article in English | MEDLINE | ID: mdl-18765157

ABSTRACT

The migration from cytokine therapy to the use of recently approved tyrosine kinase inhibitors and targeted therapeutic strategies may deprive some patients of a chance for long-term survival, as many clinicians now see these new agents as "easy fixes" for treating renal cell carcinoma (RCC). New developments pertaining to the mechanism, patient selection, predictive biomarkers, and administration of interleukin-2 mandate a reassessment of the clinical landscape and the clinical trial information upon which our current practices are based. Recalibration of the scales that we use to weigh the various possible therapies for advanced kidney cancer is also needed. Despite the shift away from cytokine therapy in the current treatment paradigm, new therapeutic approaches continue to build upon the undisputed fact that RCC can be cured with immunotherapy.


Subject(s)
Carcinoma, Renal Cell/therapy , Immunotherapy , Kidney Neoplasms/therapy , Carcinoma, Renal Cell/diagnosis , Carcinoma, Renal Cell/immunology , Clinical Trials as Topic , Humans , Kidney Neoplasms/diagnosis , Kidney Neoplasms/immunology
17.
Mol Cancer Ther ; 7(5): 1227-36, 2008 May.
Article in English | MEDLINE | ID: mdl-18483310

ABSTRACT

Plasminogen activator inhibitor-1 (PAI-1) is an important endogenous inhibitor of urokinase-type plasminogen activator. Its action in tumor angiogenesis is complicated, varying with experimental setting and its cellular origin. To further understand the mechanism of the effect of PAI-1 on tumor angiogenesis, especially newly established tumor vasculature in early tumor progression, stable transfectants (TO-PAI-1) of the human prostate adenocarcinoma, PC3, were generated in which PAI-1 expression is under the control of the tetracycline-responsive promoter (Tet-On system). The TO-PAI-1 transfectants exhibit tight inducibility of expression of biologically active PAI-1 in vitro. Induction of PAI-1 expression in nude mice resulted in significant inhibition of tumor growth. This inhibition appears to be due to the effect of PAI-1 on angiogenesis, because it is manifested by an initial wave of tumor endothelial apoptosis accompanied by induction of tumor cell apoptosis and inhibition of tumor cell proliferation. Similar endothelial apoptosis is observed in vitro when human microvascular endothelial cells are physically cocultivated with TO-PAI-1 cells on vitronectin-coated plate. Taken together, these data show for the first time that PAI-1 induces endothelial apoptosis in the newly established tumor vasculature.


Subject(s)
Apoptosis , Endothelium, Vascular/pathology , Plasminogen Activator Inhibitor 1/metabolism , Prostatic Neoplasms/blood supply , Animals , Doxycycline/pharmacology , Endothelial Cells/pathology , Humans , Male , Mice , Mice, Nude , Neovascularization, Pathologic/metabolism , Plasminogen Activator Inhibitor 1/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Tumor Cells, Cultured , Vitronectin/metabolism , Xenograft Model Antitumor Assays
18.
Int J Urol ; 14(1): 21-5, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17199855

ABSTRACT

OBJECTIVE: Epithelioid angiomyolipomas (AML) of the kidney are malignant tumors with aggressive clinical behavior. METHODS: We reviewed cases of epithelioid AML recently diagnosed at our institution to highlight the spectrum of clinical presentations. RESULTS: In all cases, renal lesions seen on computed tomography were suspicious for renal cell carcinoma (RCC). Histologically, these tumors can resemble RCC. The diagnosis of epithelioid AML was established by positive staining for melanoma and smooth muscle cell markers, and presence of perivascular epithelioid cells. One patient presented with a renal tumor extending into the inferior vena cava to the level of the hepatic veins. Two patients developed recurrent, metastatic disease following nephrectomy. One patient with tuberous sclerosis and multiple, bilateral AML developed an enhancing renal tumor that did not contain any fat densities. A partial nephrectomy was performed and pathology revealed epithelioid AML adjacent to conventional AML. CONCLUSIONS: These tumors are distinguished from RCC by positive immunostaining for melanoma markers and smooth muscle cell markers. They resemble conventional RCC on imaging. Epithelioid AML may be locally aggressive and metastasize.


Subject(s)
Angiomyolipoma/diagnosis , Kidney Neoplasms/diagnosis , Adult , Aged , Carcinoma, Renal Cell/diagnosis , Diagnosis, Differential , Female , Humans , Male , Middle Aged
19.
Proc Natl Acad Sci U S A ; 103(35): 13132-7, 2006 Aug 29.
Article in English | MEDLINE | ID: mdl-16916931

ABSTRACT

The A2A adenosine receptor (A2AR) has been shown to be a critical and nonredundant negative regulator of immune cells in protecting normal tissues from inflammatory damage. We hypothesized that A2AR also protects cancerous tissues by inhibiting incoming antitumor T lymphocytes. Here we confirm this hypothesis by showing that genetic deletion of A2AR in the host resulted in rejection of established immunogenic tumors in approximately 60% of A2AR-deficient mice with no rejection observed in control WT mice. The use of antagonists, including caffeine, or targeting the A2 receptors by siRNA pretreatment of T cells improved the inhibition of tumor growth, destruction of metastases, and prevention of neovascularization by antitumor T cells. The data suggest that effects of A2AR are T cell autonomous. The inhibition of antitumor T cells via their A2AR in the adenosine-rich tumor microenvironment may explain the paradoxical coexistence of tumors and antitumor immune cells in some cancer patients (the "Hellstrom paradox"). We propose to target the hypoxia-->adenosine-->A2AR pathway as a cancer immunotherapy strategy to prevent the inhibition of antitumor T cells in the tumor microenvironment. The same strategy may prevent the premature termination of immune response and improve the vaccine-induced development of antitumor and antiviral T cells. The observations of autoimmunity during melanoma rejection in A2AR-deficient mice suggest that A2AR in T cells is also important in preventing autoimmunity. Thus, although using the hypoxia-->adenosine-->A2AR pathway inhibitors may improve antitumor immunity, the recruitment of this pathway by selective drugs is expected to attenuate the autoimmune tissue damage.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Melanoma/immunology , Melanoma/metabolism , Receptor, Adenosine A2A/metabolism , Adenosine/metabolism , Adenosine A2 Receptor Antagonists , Animals , Cell- and Tissue-Based Therapy , Humans , Immunotherapy, Adoptive , Interferon-gamma/biosynthesis , Melanoma/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mutation/genetics , Receptor, Adenosine A2A/deficiency
20.
Curr Treat Options Oncol ; 6(6): 487-97, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16242053

ABSTRACT

Sarcomas are a heterogeneous group of tumors that respond poorly to conventional cytotoxic chemotherapy. Sarcomas possess specific molecular characteristics that exist because of unique somatic mutations, vital growth factor or growth factor receptor overexpression, or are critically dependent on host pathways. Currently these tumors are classified on the basis of their tissue of origin and histologic appearance. Gene expression and proteomic analysis of sarcomas may enable reclassification of these tumors and help predict their biologic behavior and devise common therapeutic strategies within a class. It may not be long before cDNA/protein expression profiling and karyotyping complement the current standard pathological evaluation of a newly diagnosed sarcoma, thereby helping the clinician pick targeted agent(s) relevant to the expression profile. The spectacular success of imatinib in patients with gastrointestinal stromal tumor demonstrates how molecular targeting can fulfill the promise of low toxicity and high response rates. Finding new targets, and learning how to use the current generation of targeting drugs in sarcoma, are urgent challenges. Therapeutic clinical trials with a host of new molecular-targeting agents are underway that will drive new paradigms in sarcoma therapeutics. Patients with refractory disease who currently have no viable therapeutic options may have options that open up with the advent of newer targeted approaches, converting their disease from an acute short-lived one to a chronic process with preservation of quality of life while receiving therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Design , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Sarcoma/drug therapy , Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/pharmacology , Gene Targeting , Humans , Oligonucleotides, Antisense/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , Protein Kinases/drug effects , RNA, Messenger/antagonists & inhibitors , Sarcoma/genetics , Sarcoma/pathology , TOR Serine-Threonine Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...