Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
2.
Prostate Cancer Prostatic Dis ; 18(3): 242-8, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26169045

ABSTRACT

BACKGROUND: The primary objective of this study was to compare the effects of pomegranate juice on PSA doubling times (PSADT) in subjects with rising PSA levels after primary therapy for prostate cancer. METHODS: Double-blind, placebo-controlled multi-institutional study, evaluated the effects of pomegranate liquid extract on serum PSA levels. The primary end point of this study was change in serum PSADT. Additional secondary and exploratory objectives were to evaluate the safety of pomegranate juice and to determine the interaction of manganese superoxide dismutase (MnSOD) AA genotype and pomegranate treatment on PSADT. RESULTS: One-hundred eighty-three eligible subjects were randomly assigned to the active and placebo groups with a ratio of 2:1 (extract N=102; placebo N=64; juice N=17). The majority of adverse events were of moderate or mild grade. Median PSADT increased from 11.1 months at baseline to 15.6 months in the placebo group (P<0.001) compared with an increase from 12.9 months at baseline to 14.5 months in the extract group (P=0.13) and an increase from 12.7 at baseline to 20.3 in the juice group (P=0.004). However, none of these changes were statistically significant between the three groups (P>0.05). Placebo AA patients experienced a 1.8 month change in median PSADT from 10.9 months at baseline to 12.7 months (P=0.22), while extract patients experienced a 12 month change in median PSADT from 13.6 at baseline to 25.6 months (P=0.03). CONCLUSIONS: Compared with placebo, pomegranate extract did not significantly prolong PSADT in prostate cancer patients with rising PSA after primary therapy. A significant prolongation in PSADT was observed in both the treatment and placebo arms. Men with the MnSOD AA genotype may represent a group that is more sensitive to the antiproliferative effects of pomegranate on PSADT; however, this finding requires prospective hypothesis testing and validation.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Lythraceae/chemistry , Plant Extracts/therapeutic use , Prostate-Specific Antigen/blood , Prostatic Neoplasms/blood , Prostatic Neoplasms/drug therapy , Aged , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/adverse effects , Combined Modality Therapy , Disease Progression , Humans , Male , Medication Adherence , Middle Aged , Neoplasm Grading , Neoplasm Staging , Plant Extracts/administration & dosage , Plant Extracts/adverse effects , Polymorphism, Single Nucleotide , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Superoxide Dismutase/genetics , Treatment Outcome
3.
Eur J Cancer ; 50(6): 1084-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24559686

ABSTRACT

Treatment of metastatic renal cell carcinoma (mRCC) with sunitinib is often associated with toxicity necessitating dose reduction. Maintaining adequate dosing and drug levels are essential for optimising clinical efficacy. Standard sunitinib schedule is 4 weeks of treatment and 2 weeks of rest (schedule 4/2). Empirically, several mRCC patients at The Cleveland Clinic (CCF) have been changed from schedule 4/2 to 2 weeks of treatment/1 week off (schedule 2/1) after experiencing toxicity, in an attempt to maintain daily dosing. The medical records of 30 mRCC patients on sunitinib who were changed from schedule 4/2 to schedule 2/1 at CCF were retrospectively reviewed. Toxicity on each schedule was recorded during routine clinic visits and graded using Common Toxicity Criteria, version 4.0. 97% of patients on schedule 4/2 had grade 3 or 4 toxicity that led to changing to schedule 2/1. There were no grade 4 toxicities on schedule 2/1, and 27% of patients experienced grade 3 toxicity (p=0.0001). Two of the most common toxicities, fatigue and hand-foot syndrome (HFS), were significantly less frequent on schedule 2/1 than on schedule 4/2 (p=0.0003; p=0.0004, respectively). Median overall treatment duration on schedule 4/2 was 12.6 months (range 1.2 months-5.1 years) and median overall treatment duration on schedule 2/1 was 11.9 months (range 0.9+ to 73.3+ months). Treatment with sunitinib on schedule 2/1 is associated with significantly decreased toxicity in patients who experience grade 3 or greater toxicity on schedule 4/2, and can extend treatment duration considerably. Prospective clinical trials are required to define the optimal sunitinib schedule to balance efficacy and toxicity.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Indoles/therapeutic use , Kidney Neoplasms/drug therapy , Pyrroles/therapeutic use , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/pathology , Drug Administration Schedule , Fatigue/chemically induced , Female , Hand-Foot Syndrome/etiology , Humans , Indoles/adverse effects , Kidney Neoplasms/pathology , Male , Middle Aged , Neoplasm Metastasis , Pyrroles/adverse effects , Retrospective Studies , Sunitinib , Treatment Outcome
4.
Ann Oncol ; 23(2): 406-10, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21543626

ABSTRACT

BACKGROUND: Cisplatin-based chemotherapy is a standard treatment of metastatic urothelial carcinoma (UC), though carboplatin-based chemotherapy is frequently substituted due to improved tolerability. Because comparative effectiveness in clinical outcomes of cisplatin- versus carboplatin-based chemotherapy is lacking, a meta-analysis was carried out. METHODS: PubMed was searched for articles published from 1966 to 2010. Eligible studies included prospective randomized trials evaluating cisplatin- versus carboplatin-based regimens in patients with metastatic UC. Individual patient data were not available and survival data were inconsistently reported. Therefore, the analysis focused on overall response (OR) and complete response (CR) rates. The Mantel-Haenszel method was used for combining trials and calculating pooled risk ratios (RRs). RESULTS: A total of 286 patients with metastatic UC from four randomized trials were included. Cisplatin-based chemotherapy was associated with a significantly higher likelihood of achieving a CR [RR = 3.54; 95% confidence interval (CI) 1.48-8.49; P = 0.005] and OR (RR = 1.34; 95% CI 1.04-1.71; P = 0.02). Survival end points could not be adequately assessed due to inconsistent reporting among trials. CONCLUSIONS: Cisplatin-based, as compared with carboplatin-based, chemotherapy significantly increases the likelihood of both OR and CR in patients with metastatic UC. The impact of improved response proportions on survival end points could not be assessed.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carboplatin/therapeutic use , Carcinoma, Transitional Cell/drug therapy , Cisplatin/therapeutic use , Comparative Effectiveness Research , Urologic Neoplasms/drug therapy , Carcinoma, Transitional Cell/secondary , Female , Humans , Male , Randomized Controlled Trials as Topic , Urologic Neoplasms/secondary
5.
J Urol ; 181(4): 1672-7; discussion 1677, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19230915

ABSTRACT

PURPOSE: Taxane based chemotherapy has activity in advanced prostate cancer but previous studies of neoadjuvant docetaxel demonstrated a prostate specific antigen response with no obvious antitumor activity. The efficacy and safety of neoadjuvant albumin-bound paclitaxel (nab-paclitaxel, Abraxane), a novel nanoparticle based formulation, were assessed in patients with high risk, locally advanced prostate cancer. MATERIALS AND METHODS: Eligible patients had locally advanced prostatic adenocarcinoma, clinical stage cT2b or greater, Gleason score 8 or greater, or serum prostate specific antigen 15 ng/ml or greater without metastatic disease. Patients received 2 cycles of 150 mg/m(2) nab-paclitaxel weekly for 3 weeks during each 4-week cycle, followed by radical prostatectomy with bilateral lymphadenectomy. Efficacy assessments included pathological and prostate specific antigen response. RESULTS: A total of 19 patients completed neoadjuvant therapy and 18 underwent radical prostatectomy. Median pretreatment prostate specific antigen was 8.5 ng/ml and median Gleason score was 8. Despite the lack of complete pathological responses 5 of 18 patients (28%) had organ confined disease and 9 of 18 (50%) had specimen confined disease. Post-chemotherapy prostate specific antigen was decreased in 18 of 19 (95%) patients and median decrease was 2.9 ng/ml (35%, p <0.001). An initial prostate specific antigen after radical prostatectomy of 0.02 ng/ml or less was achieved in 17 of 18 (94%) patients. There were no significant perioperative complications. Cytoplasmic vacuolization (focal in 10 and extensive in 7) was evident in all but 1 patient (94%). Ten patients (56%) had grade 3 and 1 had grade 4 neutropenia with no febrile neutropenia. CONCLUSIONS: Neoadjuvant nab-paclitaxel was well tolerated. Similar to our experience with neoadjuvant docetaxel there were no pathological complete responses, although a possible histological antitumor effect was observed.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/surgery , Albumins/therapeutic use , Paclitaxel/therapeutic use , Prostatectomy , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/surgery , Aged , Humans , Male , Middle Aged , Neoadjuvant Therapy , Risk Factors
6.
Invest New Drugs ; 27(4): 379-86, 2009 Aug.
Article in English | MEDLINE | ID: mdl-18931824

ABSTRACT

PURPOSE: TG4010 is a recombinant MVA vector expressing the tumor-associated antigen MUC1 and IL2. We explored the effect two schedules of TG4010 on PSA in men with PSA progression. PATIENTS AND METHODS: A randomized phase II trial was conducted in 40 patients with PSA progression. Patients had PSA doubling times less than 10 months, with no overt evidence of disease. Patients received either weekly subcutaneous injection (sc) of TG4010 10(8) pfu for 6 weeks, then one injection every 3 weeks or sc injection of TG4010 10(8) pfu every 3 weeks. RESULTS: The primary endpoint of a 50% decrease in PSA values from baseline was not observed. Nevertheless, 13 of 40 patients had a more than two fold improvement in PSA doubling time. Ten patients had their PSA stabilized for over 8 months. Therapy was well tolerated. CONCLUSIONS: Although the primary endpoint was not achieved, there is evidence of biologic activity of TG4010 in patients with PSA progression, further investigation in prostate cancer is warranted.


Subject(s)
Cancer Vaccines/therapeutic use , Membrane Glycoproteins/therapeutic use , Prostate-Specific Antigen/drug effects , Prostatic Neoplasms/drug therapy , Aged , Cancer Vaccines/administration & dosage , Cancer Vaccines/adverse effects , Disease Progression , Drug Administration Schedule , Humans , Immunotherapy/methods , Injections, Subcutaneous , Interleukin-2/immunology , Male , Membrane Glycoproteins/administration & dosage , Membrane Glycoproteins/adverse effects , Middle Aged , Mucin-1/immunology , Prostate-Specific Antigen/blood , Prostatic Neoplasms/immunology , Time Factors , Treatment Outcome
7.
Invest New Drugs ; 26(6): 553-9, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18626572

ABSTRACT

UNLABELLED: Docetaxel is second generation taxoid that has shown activity against a variety of cancers and has been approved for use in cancers of the breast, lung, head and neck, ovaries and prostate. Temozolomide is an alkylating agent which crosses the blood brain barrier and has demonstrated antitumor activity against a broad range of tumor types, including malignant glioma, melanoma, non small cell lung cancer and carcinoma of the ovary and colon. A Phase I trial was conducted to determine the toxicity of this combination in refractory solid tumor patients. METHODS: Twenty five patients with metastatic cancers were enrolled in a Phase I dose escalation trial. Docetaxel was administered weekly in 5 escalating doses of 25 to 35 mg/ m(2) as a one-hour bolus intravenous infusion for 3 consecutive weeks. Temozolamide was administered orally daily for 3 weeks (escalating doses of 75 to 100 mg/m(2)). Cycles were repeated at 4 week intervals. RESULTS: The maximum tolerated dose (MTD) was not determined in this study. The most commonly reported adverse events were mild to moderate nausea, vomiting and fatigue. Thrombocytopenia was the most commonly observed grade 3 and 4 hematological toxicity. Eight patients had dose interruptions for adverse events and only one patient had a dose reduction while receiving 30 mg/ m(2) of docetaxel and 90 mg/ m(2) of temozolomide due to grade 3 thrombocytopenia. Two patients achieved partial responses and 88% of the patients are deceased. The median survival is 8.4 months. CONCLUSIONS: The combination of docetaxel and temozolomide was well tolerated and these agents can be safely combined. For phase II trials, docetaxel 35 mg/ m(2) IV day 1, 8 and 15, and daily temozolomide at 100 mg/ m(2) day 1-21 are recommended.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasms/drug therapy , Administration, Oral , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Docetaxel , Dose-Response Relationship, Drug , Female , Humans , Infusions, Intravenous , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Metastasis , Survival Rate , Taxoids/administration & dosage , Temozolomide
8.
Ann Oncol ; 19(2): 265-8, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17962201

ABSTRACT

BACKGROUND: Sorafenib is an orally bioavailable vascular endothelial growth factor receptor (VEGFR) inhibitor with antitumor activity in metastatic renal cell carcinoma (RCC). Sunitinib, also a VEGFR inhibitor, induces biochemical hypothyroidism in 85% of metastatic RCC patients, the majority of whom have signs or symptoms of hypothyroidism. Hence, the incidence of thyroid function test (TFT) abnormalities in patients with metastatic RCC receiving sorafenib was investigated. PATIENTS AND METHODS: Sixty-eight patients with metastatic RCC were treated with sorafenib at the Cleveland Clinic Taussig Cancer Center, and 39 patients had TFTs available. RESULTS: Eight patients (21%) had thyroid dysfunction possibly caused by sorafenib [seven hypothyroidism (18%) and one hyperthyroidism (3%)] and eight additional patients (21%) had findings compatible with nonthyroidal illness. Only two patients had clinical signs and symptoms secondary to thyroid dysfunction and received thyroid hormone replacement. CONCLUSIONS: In summary, clinically significant TFT abnormalities were not common in patients treated with sorafenib, and replacement therapy was rarely indicated. TFTs should be measured before sorafenib therapy in RCC patients and subsequently only if clinically indicated.


Subject(s)
Benzenesulfonates/adverse effects , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/secondary , Hypothyroidism/chemically induced , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Pyridines/adverse effects , Administration, Oral , Adult , Aged , Aged, 80 and over , Benzenesulfonates/therapeutic use , Carcinoma, Renal Cell/pathology , Chemotherapy, Adjuvant , Cohort Studies , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Follow-Up Studies , Humans , Hypothyroidism/epidemiology , Kidney Neoplasms/mortality , Male , Middle Aged , Neoplasm Metastasis , Niacinamide/analogs & derivatives , Phenylurea Compounds , Pyridines/therapeutic use , Retrospective Studies , Risk Assessment , Sorafenib , Thyroid Function Tests
9.
Invest New Drugs ; 25(3): 211-6, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17103043

ABSTRACT

INTRODUCTION: Lenalidomide is an immunomodulatory derivative of thalidomide with significantly greater in vitro activity and a different toxicity profile. In preclinical trials it has shown synergy with chemotherapy. PATIENTS AND METHODS: Primary objective of this study was to determine the maximum tolerated doses of docetaxel and carboplatin when combined with oral lenalidomide in a standard phase I study design. Between September 2004 and May 2005, 14 patients with pathologically proven solid tumors, < or =2 prior chemotherapy regimens, performance status ECOG 0/1, and adequate organ function were enrolled. Dose limiting toxicities (DLT) were defined as > or = grade 3 non-hematological, or grade 4 hematological toxicity. No growth factors were used during cycle 1. RESULTS: Three of four patients treated at dose level 1, docetaxel 60 mg/m(2) and carboplatin AUC 6 on Day 1, and lenalidomide 10 mg orally daily on Days 1-14 of a 21 day cycle experienced DLT (grade 3 electrolyte changes in two patients, and grade 4 neutropenia in one patient). Ten patients were treated at dose level -1, docetaxel 60 mg/m(2) and carboplatin AUC 6 on Day 1, and lenalidomide 5 mg orally daily on Days 1-14 of a 21 day cycle with one DLT (Grade 4 neutropenia). There were no treatment-related deaths or irreversible toxicities. Of the 14 response-evaluable patients, five achieved a partial response (5 out of 9 patients with non-small cell lung cancer. CONCLUSIONS: Docetaxel 60 mg/m(2) and carboplatin AUC 6 on Day 1, with lenalidomide 5 mg orally daily on Days 1-14 days of a 21 day cycle is the maximum tolerated dose without the use of prophylactic growth factors. This combination is active and further evaluation in a phase II trial is warranted.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasms/drug therapy , Administration, Oral , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carboplatin/administration & dosage , Docetaxel , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Infusions, Intravenous , Lenalidomide , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/pathology , Taxoids/administration & dosage , Thalidomide/administration & dosage , Thalidomide/analogs & derivatives , Treatment Outcome
10.
Semin Oncol ; 28(4 Suppl 15): 45-8, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11685728

ABSTRACT

Patients with locally advanced prostate cancer present a significant therapeutic dilemma. Despite aggressive local therapies, including radical prostatectomy (RP), these patients are at high risk for biochemical failure. Several research groups have recently demonstrated the feasibility of hormonal and chemohormonal therapy before RP, but limited published data are available regarding the usefulness of chemotherapy without hormonal therapy in the neoadjuvant setting. At Cleveland Clinic Foundation, a phase II trial was initiated to evaluate a 6-week course of docetaxel, 40 mg/m(2) intravenously every 7 days, followed by RP in patients with locally advanced prostate cancer. RP was to be performed within 3 weeks of completion of neoadjuvant chemotherapy. The primary endpoint of this study is pathologic complete response. Preliminary toxicity data suggest that weekly docetaxel is well tolerated and does not increase the risk of perioperative or post operative complications. Reductions in prostate-specific antigen levels were noted in seven of 10 patients who completed the 6-week course of neoadjuvant docetaxel. The neoadjuvant use of investigational cancer therapies may allow for relatively rapid assessment of their antitumor activity.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Paclitaxel/analogs & derivatives , Paclitaxel/therapeutic use , Prostatic Neoplasms/drug therapy , Taxoids , Aged , Docetaxel , Humans , Male , Middle Aged , Neoadjuvant Therapy , Prostatectomy , Prostatic Neoplasms/surgery
11.
Invest New Drugs ; 19(3): 261-5, 2001.
Article in English | MEDLINE | ID: mdl-11561685

ABSTRACT

OBJECTIVES: Prostate-specific antigen only disease progression following definitive therapy is significant therapeutic dilemma. The benefit of hormonal therapy remains unproven and is associated with significant toxicity, more pronounced with chronic use. Biochemical progression following hormonal therapy has no standard treatment. New approaches to the management of this subset of patients are needed. A previous study in advanced prostate cancer demonstrated biologic activity of granulocyte macrophage-colony stimulating factor. The purpose of this study was to evaluate the activity of granulocyte macrophage-colony stimulating factor in a less heavily pretreated population. MATERIALS AND METHODS: Sixteen patients with advanced prostate cancer, 7 hormonally naïve, and 9 androgen independent, were treated with granulocyte macrophage-colony stimulating factor administered subcutaneously at 250 microg three times a week for up to 6 months. Prostate-specific antigen measurements were obtained every 2 weeks. RESULTS: No patient achieved an objective response. Six patients demonstrated a 10-15% decline in their baseline prostate-specific antigen which was maintained during the entire treatment period. Five of these 6 patients demonstrated a rise in their prostate-specific antigen following study completion. Therapy was well tolerated, with only 1 grade 3 event which was not treatment-related. CONCLUSIONS: Granulocyte macrophage-colony stimulating factor demonstrates modest biologic evidence of activity in prostate cancer as manifested by prostate-specific antigen response. Further investigation of the mechanism of activity and additional clinical evaluation of this agent seems warranted.


Subject(s)
Adenocarcinoma/drug therapy , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Prostatic Neoplasms/drug therapy , Aged , Granulocyte-Macrophage Colony-Stimulating Factor/adverse effects , Humans , Male , Middle Aged , Prostate-Specific Antigen/blood , Recombinant Proteins , Treatment Outcome
12.
Semin Urol Oncol ; 19(3): 180-5, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11561985

ABSTRACT

Muscle-invasive bladder cancer is typically an aggressive solid tumor with the propensity for early systemic dissemination. Although radical cystectomy remains the gold standard intervention, the high rate of systemic failure has prompted investigators to evaluate various strategies to attempt to improve survival, including the early administration of systemic chemotherapy. These efforts have provided mixed results with two recently completed trials providing conflicting results. Other strategies include attempts to both preserve the bladder using combinations of limited surgical resection, systemic chemotherapy, and radiotherapy. This review focuses on the potential of neoadjuvantly administered therapies to impact the management of muscle-invasive bladder cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Urinary Bladder Neoplasms/drug therapy , Chemotherapy, Adjuvant/methods , Cisplatin/therapeutic use , Cystectomy/methods , Follow-Up Studies , Humans , Neoplasm Invasiveness , Randomized Controlled Trials as Topic , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/radiotherapy , Urinary Bladder Neoplasms/surgery
13.
Cancer ; 91(7): 1191-200, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11283917

ABSTRACT

BACKGROUND: This study evaluated the dose-response relation for zoledronic acid, a new generation high potency bisphosphonate, given as a 5-minute infusion in patients with malignant osteolytic disease. METHODS: Two-hundred eighty patients with osteolytic lesions due to metastatic breast carcinoma or multiple myeloma were randomized to double-blind treatment with either 0.4, 2.0, or 4.0 mg of zoledronic acid or 90 mg pamidronate. The primary efficacy endpoint was the proportion of patients receiving radiation to bone. Other skeletal-related events, bone mineral density (BMD), bone markers, Eastern Cooperative Oncology Group performance status, pain and analgesic scores, and safety also were evaluated. RESULTS: Zoledronic acid at doses of 2.0 and 4.0 mg and pamidronate at a dose of 90 mg each significantly reduced the need for radiation therapy to bone (P < 0.05) in contrast with 0.4 mg zoledronic acid, which did not. Skeletal-related events of any kind, pathologic fractures, and hypercalcemia also occurred less frequently in patients treated with 2.0 or 4.0 mg zoledronic acid or pamidronate than with 0.4 mg zoledronic acid. Increases in lumbar spine BMD (6.2-9.6%) and decreases in the bone resorption marker N-telopeptide (range, -37.1 to -60.8%) were observed for all treatment groups. Skeletal pain, fatigue, nausea, vomiting, and headache were the most commonly reported adverse events. Adverse events were similar in nature and frequency with zoledronic acid and pamidronate. CONCLUSIONS: A 5-minute infusion of 2.0-4.0 mg zoledronic acid was at least as effective as a 2-hour 90-mg pamidronate infusion in treatment of osteolytic metastases. A 0.4-mg dose of zoledronic acid was significantly less effective. Both zoledronic acid and pamidronate were well tolerated.


Subject(s)
Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Diphosphonates/administration & dosage , Imidazoles/administration & dosage , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Biomarkers, Tumor/urine , Bone Density/drug effects , Bone Neoplasms/complications , Bone Neoplasms/radiotherapy , Bone Resorption , Breast Neoplasms/pathology , Collagen/urine , Collagen Type I , Creatinine/urine , Diphosphonates/adverse effects , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Imidazoles/adverse effects , Infusions, Intravenous , Male , Middle Aged , Multiple Myeloma/pathology , Pain Measurement , Pamidronate , Peptides/urine , Zoledronic Acid
14.
Urology ; 57(2): 281-5, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11182337

ABSTRACT

OBJECTIVES: To report the results of a Phase II trial of neoadjuvant estramustine and etoposide before radical prostatectomy in patients with locally advanced disease. METHODS: Treatment consisted of three cycles of estramustine (10 mg/kg/day) and etoposide (50 mg/m(2)/day) orally on days 1 through 21, repeated every 28 days, followed by radical prostatectomy. The eligibility criteria included locally advanced prostate cancer (clinical Stage T2b/c or T3, prostate-specific antigen [PSA] level of 15 ng/mL or greater, or Gleason score of 8 or higher) without evidence of metastatic disease. The median PSA level was 14 ng/mL (range 5.3 to 50), the median Gleason score was 7 (range 6 to 9), and 44% had Stage T2b/c or T3 disease. The primary endpoint was feasibility of neoadjuvant therapy and radical prostatectomy, including drug and surgery-related toxicities. Secondary endpoints included the pre-prostatectomy PSA level, local response, pathologic outcomes, and time to PSA failure. RESULTS: Eighteen patients were entered and completed all three cycles of therapy, and 16 (89%) underwent radical prostatectomy. A local response occurred in 15 (94%) of 16 patients with palpable tumors, and the serum PSA reached undetectable levels after therapy and before radical prostatectomy in 9 patients (50%). Five patients (28%) experienced grade 3 toxicity (two with deep venous thrombosis, two with neutropenia, and one with diarrhea) and one (6%) experienced grade 4 toxicity (pulmonary embolus) before surgery. The median operative time was 125 minutes, the mean blood loss was 665 mL, and the mean length of stay was 2.5 nights. Five minor surgical complications occurred in 4 patients. The pathologic analysis demonstrated residual carcinoma with squamous metaplasia and androgen deprivation effect in all patients. Five patients (31%) had organ-confined disease and 9 patients (56%) had specimen-confined disease. All patients achieved an undetectable PSA level postoperatively and at a median follow-up of 14 months (range 5 to 20) and without additional therapy, all 14 patients with negative lymph nodes were disease free. CONCLUSIONS: This trial confirms the feasibility of radical prostatectomy with acceptable surgical morbidity after neoadjuvant therapy with estramustine and etoposide in patients with locally advanced prostate cancer. However, this regimen is associated with estramustine-induced thromboembolic toxicity. The results of the pathologic analysis suggest a higher than expected rate of organ-confined and specimen-confined disease, but little histologic evidence of antitumor effect beyond that associated with androgen deprivation. Additional study of this paradigm with other drug regimens is warranted.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Estramustine/therapeutic use , Etoposide/therapeutic use , Prostatectomy/methods , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/surgery , Antineoplastic Agents, Hormonal/adverse effects , Antineoplastic Agents, Phytogenic/adverse effects , Estramustine/adverse effects , Etoposide/adverse effects , Follow-Up Studies , Humans , Male , Middle Aged , Neoadjuvant Therapy , Prostate-Specific Antigen/blood , Prostatic Neoplasms/pathology , Treatment Outcome
15.
Curr Treat Options Oncol ; 2(5): 403-11, 2001 Oct.
Article in English | MEDLINE | ID: mdl-12057103

ABSTRACT

Standard therapy for clinically localized prostate cancer includes radical prostatectomy, external beam radiotherapy, or transperineal interstitial brachytherapy. Patients eligible for standard therapy are those with low risk features as defined by various risk group classifications, which generally include clinical stage T1 or T2a, serum prostate-specific antigen (PSA) less than 10 ng/mL, and biopsy Gleason sum of 6 or less. Although there has been important evolution in the performance of these techniques, particularly with respect to functional outcomes, these approaches for low-risk disease are relatively mature, and the cure rates with each of these therapies are similar in this patient population; locally advanced disease is more difficult to cure, however. Biochemical disease-free survival rates in men undergoing radical prostatectomy are clearly related to the pathologic stage. Prognostic groups can be defined based on pathologic stage with increasingly worse outcomes based on extracapsular extension (ECE), margin status, and the status of the lymph nodes and seminal vesicles. In patients with low risk features, the positive margin rate is generally low, making the presence or absence of ECE the dominant variable in predicting the likelihood of treatment failure. These observations suggest that more aggressive therapy is needed to cure those who are likely to have ECE or other adverse histologic features. Several nomograms predicting the likelihood of ECE or 5-year biochemical failure rates are now in routine clinical use, and can be used to select men at high risk of failure with single modality therapy for more aggressive treatment strategies. However, the optimal form of aggressive therapy for these patients is unknown.


Subject(s)
Adenocarcinoma/pathology , Prostatic Neoplasms/pathology , Adenocarcinoma/epidemiology , Adenocarcinoma/therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brachytherapy , Chemotherapy, Adjuvant , Combined Modality Therapy , Disease-Free Survival , Epidemiologic Methods , Humans , Male , Neoadjuvant Therapy , Neoplasm Invasiveness , Neoplasm Staging , Prognosis , Prostatectomy , Prostatic Neoplasms/epidemiology , Prostatic Neoplasms/therapy , Radiotherapy, Adjuvant , Radiotherapy, Conformal , Risk Factors , Treatment Outcome
16.
Curr Treat Options Oncol ; 2(5): 431-6, 2001 Oct.
Article in English | MEDLINE | ID: mdl-12057106

ABSTRACT

Metastatic transitional cell carcinoma of the bladder is an aggressive neoplasm characterized by rapid growth and dissemination with a median survival of typically less than 1 year. Despite the availability of a myriad of antineoplastics with moderate-significant anti-tumor activity yielding overall response rates in the 40% to 80% range, randomized trials continue to demonstrate median survival rates in the 13- to14-month range, with very limited long-term survival. Subsets of patients with advanced bladder cancer present additional management problems, including those with renal insufficiency or nontransitional-cell histology. Various observers have noted the similarity in treatment outcomes in advanced bladder cancer and extensive small cell lung cancer where chemotherapy produces relatively high response rates but with limited impact on survival. The optimal chemotherapy combination for patients with advanced bladder cancer remains undefined, however, there is increasing recognition that in order to achieve tangible improvements in complete response rates and survival in this disease will likely require a combination of chemotherapy and targeted molecular therapies and in some settings adjunctive surgery.


Subject(s)
Carcinoma, Transitional Cell/pathology , Urinary Bladder Neoplasms/pathology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma/mortality , Carcinoma/pathology , Carcinoma/therapy , Carcinoma, Transitional Cell/mortality , Carcinoma, Transitional Cell/therapy , Cisplatin/administration & dosage , Cisplatin/adverse effects , Combined Modality Therapy , Cystectomy , Epidemiologic Methods , Forecasting , Humans , Kidney Failure, Chronic/complications , Neoplasm Invasiveness , Neoplasm Metastasis , Prognosis , Survival Rate , Treatment Outcome , Urinary Bladder Neoplasms/mortality , Urinary Bladder Neoplasms/therapy
17.
Cleve Clin J Med ; 67(10): 720-2, 725-6, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11060958

ABSTRACT

Earlier diagnosis and treatment of prostate cancer has changed the face of late-stage disease, and the use of mainstay hormonal therapies--orchiectomy, luteinizing hormone releasing-hormone analogs, and combined androgen ablation--are evolving rapidly. New approaches such as antiandrogen monotherapy and intermittent therapy are being evaluated. In addition, palliative treatments for patients with androgen-independent tumors have expanded. The most common clinical presentation of advanced prostate cancer is a rising prostate-specific antigen level following primary therapy (radical prostatectomy or radiotherapy or both). Due to the negative psychological implications of orchiectomy, many patients are opting for treatment with luteinizing hormone-releasing hormone analogs. Because studies of combined androgen ablation have not provided conclusive results, it is reasonable to forego antiandrogen therapy for patients who undergo bilateral orchiectomy. Management options for patients with androgen-independent prostate cancer are expanding and include antiandrogen removal, antiandrogen therapy, and glucocorticoids.


Subject(s)
Androgen Antagonists/therapeutic use , Antineoplastic Agents, Hormonal/therapeutic use , Neoplasm Recurrence, Local/drug therapy , Prostatic Neoplasms/drug therapy , Biomarkers, Tumor/blood , Disease-Free Survival , Glucocorticoids/therapeutic use , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Hot Flashes/prevention & control , Humans , Male , Neoplasm, Residual , Orchiectomy , Palliative Care , Prostate-Specific Antigen/blood , Prostatic Neoplasms/pathology , Pulse Therapy, Drug , Randomized Controlled Trials as Topic
18.
Semin Cancer Biol ; 10(2): 125-44, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10936063

ABSTRACT

IFNs were the first new therapeutic products resulting from recombinant DNA technology. IFNs were also the first human proteins effective in cancer treatment. There is however much to be discovered which will lead to new clinical applications. Areas which represent major research challenges for full understanding and application of the IFN system are: (i) the diversity of the IFN family; (ii) the role of induction; (iii) molecular mechanism of action; (iv) cellular modulatory effects; (v) advantages of combinations, and (vi) identification of new therapeutic indications. This review will emphasize the diversity of the IFN family and chemical modifications which will result in second-generation IFNs. Pre-clinical and clinical findings form the basis for new therapeutic directions in chronic myelogenous leukemia, lymphomas, myelomas, melanoma, urologic malignancies, primary brain tumors, and ovarian carcinoma.


Subject(s)
Antineoplastic Agents/pharmacology , Interferon Type I/pharmacology , Interferon-gamma/pharmacology , Neoplasms/drug therapy , Animals , Humans , Recombinant Proteins
19.
J Clin Oncol ; 18(5): 1058-61, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10694557

ABSTRACT

PURPOSE: Cisplatin and paclitaxel are active agents in advanced urothelial cancer. A phase II trial of this combination was performed to determine the activity and toxicity of these agents in a multi-institutional setting. PATIENTS AND METHODS: Fifty-two patients with advanced urothelial carcinoma were treated on one day with paclitaxel 175 mg/m(2) over 3 hours followed by cisplatin 75 mg/m(2), both intravenously, every 21 days. Cycles were repeated every 21 days until progression or a maximum of six cycles. RESULTS: Twenty-six patients obtained an objective response, for an overall response rate of 50% (95% confidence interval, 36% to 64%). Four patients achieved complete clinical responses. The median overall survival time for the group was 10.6 months. Toxicity was moderate, with granulocytopenia and neurotoxicity being the most common side effects noted. CONCLUSION: The combination of cisplatin and paclitaxel is active in advanced urothelial cancer. Responses in visceral, nodal, and soft tissues sites were observed. Granulocytopenia without fever and grade 2/3 neurotoxicity were common. The confidence interval of the overall response rate in this study overlaps most of the other reported regimens. The optimal therapy for advanced urothelial cancer remains undefined.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Transitional Cell/drug therapy , Cisplatin/therapeutic use , Paclitaxel/therapeutic use , Urologic Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cisplatin/administration & dosage , Cisplatin/adverse effects , Female , Humans , Male , Middle Aged , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Treatment Outcome , Urothelium
20.
Invest New Drugs ; 17(2): 183-6, 1999.
Article in English | MEDLINE | ID: mdl-10638490

ABSTRACT

This study was conducted to assess the efficacy and toxicity of suramin administered using a fixed dose schedule in patients with advanced renal cell carcinoma. Fourteen eligible patients with advanced renal cell carcinoma were enrolled and treated on a fixed dose schedule of suramin administered over 12 weeks. Suramin was administered by intravenous infusions over 1 hour. None of the 13 evaluable patients demonstrated an objective response. Only 3 patients completed the 12-week therapy course, with the majority developing progressive disease on therapy. The fixed dosage schedule was well tolerated with minimal to moderate toxicity. Suramin in this fixed dose schedule is well tolerated but has no activity in advanced renal cell carcinoma.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Suramin/administration & dosage , Suramin/adverse effects , Adult , Aged , Carcinoma, Renal Cell/pathology , Drug Administration Schedule , Female , Humans , Infusions, Intravenous , Kidney Neoplasms/pathology , Male , Middle Aged , Neoplasm Metastasis/drug therapy , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...