51
|
Vestermark LW, Larsen S, Lindeløv B, Bastholt L. A phase II study of thalidomide in patients with brain metastases from malignant melanoma. Acta Oncol 2008; 47:1526-30. [PMID: 18607876 DOI: 10.1080/02841860801918521] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Brain metastases develop in nearly half of the patients with advanced melanoma and in 15 to 20% of these patients CNS is the first site of relapse. Overall median survival is short, ranging from 2 to 4 months. Thalidomide has antiangiogenic and immunomodulatory effects. Results obtained in prior trials indicate that Thalidomide acts as a cytostatic agent in metastatic melanoma. We evaluated single agent antitumour activity and toxicity of Thalidomide in a phase II setting in patients with brain metastases associated with metastatic melanoma. MATERIAL AND METHODS Patients with measurable metastatic melanoma in progression and with PS < or = 2 were enrolled in the study. Thalidomide was given orally. Dose was escalated over 4 weeks from 100 mg/day to 400 mg/day. Primary objective of the study was to determine response rate, according to RECIST. Secondary objectives were to estimate time to progression, overall survival and to evaluate tolerability of the regimen. RESULTS Twenty five men and 11 women were enrolled in the study, median age 48 years. Among 36 eligible patients 35 were evaluable for response. None of the patients obtained a response in brain metastases. Three patients obtained a partial response in extracranial lesions. Toxicity was acceptable and manageably. Median time to progression and overall survival time was 1.7 and 3.1 months, respectively. CONCLUSION There were no objective responses in the brain but single agent Thalidomide has some activity in melanoma patients with brain metastases. It has encouraged us to investigate Thalidomide in combination with Temozolomide, a very lipophilic agent, in this group of patients.
Collapse
|
52
|
Yau T, Chan P, Epstein R, Poon RT. Evolution of systemic therapy of advanced hepatocellular carcinoma. World J Gastroenterol 2008; 14:6437-41. [PMID: 19030192 PMCID: PMC2773326 DOI: 10.3748/wjg.14.6437] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) commonly occurs in hepatitis B endemic areas, especially in Asian countries. HCC is highly refractory to cytotoxic chemotherapy. This resistance is partly related to its tumor biology, pharmacokinetic properties, and both intrinsic and acquired drug resistance. There is no convincing evidence thus far that systemic chemotherapy improves overall survival in advanced HCC patients. Other systemic approaches, such as hormonal therapy and immunotherapy, have also disappointing results. Recently, encouraging results have been shown in using sorafenib in the treatment of advanced HCC patients. In this review, we concisely summarize the evolution of developments in the systemic therapy of advanced HCC.
Collapse
|
53
|
Seadi Pereira PJ, Noronha Dornelles F, Santiago Santos D, Batista Calixto J, Bueno Morrone F, Campos MM. Nociceptive and inflammatory responses induced by formalin in the orofacial region of rats: effect of anti-TNFalpha strategies. Int Immunopharmacol 2008; 9:80-5. [PMID: 18957334 DOI: 10.1016/j.intimp.2008.10.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2008] [Revised: 09/30/2008] [Accepted: 10/03/2008] [Indexed: 01/19/2023]
Abstract
This study evaluated the effects of different anti-TNFalpha strategies on the nociceptive and inflammatory responses triggered by formalin in the rat orofacial region. Formalin injection (2.5%) into the right upper lip caused a nociceptive response that was biphasic, with the first phase observed between 0 and 3 min and the second phase between 12 and 30 min. Plasma extravasation induced by formalin was time-related and reached the peak at 360 min. The monoclonal antibody anti-TNFalpha (25 and 50 pg/lip) significantly inhibited the second phase of formalin-induced nociceptive behavior, while the first phase remained unaltered. The systemic treatment with the chimeric anti-TNFalpha antibody infliximab also caused a significant inhibition of the second phase. Interestingly, the local administration of infliximab (50 pg/lip) produced a significant reduction of both phases of formalin-induced nociception. In addition, the systemic pretreatment with the preferential inhibitor of TNFalpha synthesis thalidomide (25 and 50 mg/kg, p.o) promoted a marked reduction of the first and second phases of formalin-evoked nociception. The local administration of the monoclonal antibody anti-TNFalpha (25 and 50 pg/lip) or infliximab (50 pg/lip) markedly reduced the plasma extravasation induced by formalin. Otherwise, formalin-elicited plasma extravasation was not significantly affected by the systemic administration of either infliximab (1 mg/kg; s.c) or thalidomide (50 mg/kg, p.o). Present data suggest that blocking TNFalpha effects, through different pharmacological tools, could represent a good alternative to control orofacial inflammatory pain that is refractory to other drugs.
Collapse
|
54
|
The G-rich promoter and G-rich coding sequence of basic fibroblast growth factor are the targets of thalidomide in glioma. Mol Cancer Ther 2008; 7:2405-14. [DOI: 10.1158/1535-7163.mct-07-2398] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
55
|
Bowcock SJ, Minchom A, Yates LR, Ryali MM. Ultra low dose thalidomide in elderly patients with myeloma. Br J Haematol 2008; 141:120-2. [DOI: 10.1111/j.1365-2141.2008.06989.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
56
|
|
57
|
Hutchins LF, Moon J, Clark JI, Thompson JA, Lange MK, Flaherty LE, Sondak VK. Evaluation of interferon alpha-2B and thalidomide in patients with disseminated malignant melanoma, phase 2, SWOG 0026. Cancer 2008; 110:2269-75. [PMID: 17932881 DOI: 10.1002/cncr.23035] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Southwest Oncology Group protocol 0026 evaluated interferon alpha-2b plus thalidomide in patients with disseminated melanoma. Endpoints were 6-month progression-free survival rate, response rate, and toxicity. METHODS Twenty-six patients with Stage IV melanoma, measurable or nonmeasurable disease, performance status of 0-2, and adequate renal and hepatic functions were registered. One prior systemic therapy for Stage IV disease was required. Interferon was administered subcutaneously (1 million U) twice daily; thalidomide was orally administered (200-400 mg) each evening in a dose-escalating manner. Response evaluations using Response Evaluation Criteria in Solid Tumors were performed every 8 weeks. RESULTS After 2 sudden deaths and 1 grade 4 treatment-related pulmonary embolism, this study was temporarily closed. One patient with deep-vein thrombosis and 2 with grade 3 cardiac arrhythmias were reported. The relationship of these events to the treatment was worrisome but not definitive. Grade 3 treatment-related adverse events occurred in 14 of 26 patients. Because of concern for patient safety the study was permanently closed. No treatment responses were seen in the 22 evaluable patients. Estimated 6-month progression-free survival rate was 15% (95% confidence interval [CI], 2%-29%), estimated 6-month overall survival was 58% (95% CI, 39%-77%), and estimated response probability was 0 of 22 (95% CI, 0%-15%). CONCLUSIONS This regimen demonstrated a lack of response and was associated with multiple severe toxicities. Further investigation of interferon alpha-2b and thalidomide in this dose and schedule is not warranted.
Collapse
Affiliation(s)
- Laura F Hutchins
- Division of Hematology/Oncology, Arkansas Cancer Research Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | | | | | | | | | | | | |
Collapse
|
58
|
Liu KH, Liao LM, Ro LS, Wu YL, Yeh TS. Thalidomide attenuates tumor growth and preserves fast-twitch skeletal muscle fibers in cholangiocarcinoma rats. Surgery 2007; 143:375-83. [PMID: 18291259 DOI: 10.1016/j.surg.2007.09.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2007] [Revised: 09/06/2007] [Accepted: 09/14/2007] [Indexed: 10/22/2022]
Abstract
BACKGROUND The prognosis for cholangiocarcinoma remains dismal due to a low resection rate and early recurrence. Cancer cachexia is associated with decreased survival and poor quality of life. Herein, we present a rat model of cholangiocarcinoma and demonstrate that thalidomide attenuates tumor growth and improves cachexia. METHODS A cholangiocarcinoma model was established using Sprague-Dawley rats that were fed thioacetamide for 40 weeks. Cholangiocarcinoma rats were treated using either thalidomide or saline for 8 weeks. Tumor growth and body weight were recorded for all animals. The expression of CD31, VEGF, and eIF4E of cholangiocarcinoma were determined using immunohistochemistry. Level of apoptosis and Fas-mediated apoptosis genes of cholangiocarcinoma were determined using TUNEL assay and ribonuclease protection assay, respectively. The distribution of fast-twitch soleus skeletal muscle fibers was determined as was the expression of TNFalpha and TGFbeta1 within soleus muscle. RESULTS After an 8-week treatment, the mean weight of saline- and thalidomide-treated rats was 24% and 19%, respectively, less than that of control (ANOVA, P < .05). The tumor volume (x +/-SD) of thalidomide-treated rats was less than saline-treated rats (1.9 +/- 0.4 vs 4.6 +/- 1.3 cm3, P < .01). The expression of CD31, eIF4E, and VEGF of cholangiocarcinoma was less than thalidomide-treated rats than for saline-treated rats, while the level of apoptosis of tumor cells was greater for thalidomide- treated rats than for saline-treated rats. The expression of mRNA for Fas, caspase-3, and Bax of cholangiocarcinoma in the thalidomide-treated rats was greater than for saline-treated rats. The number of fast-twitch skeletal muscle fibers per 500 mm2 of control, saline-, and thalidomide-treated rats was 43 +/- 6, 14 +/- 3, and 41 +/- 8 (ANOVA, P < .001). The expression of TNFalpha and TGFbeta1 of soleus muscles for thalidomide-treated rats was less than for saline-treated rats. CONCLUSIONS Using our rat cholangiocarcinoma model, we demonstrated that thalidomide inhibited tumor growth and was associated with a decrease in expression of reduced eIF4E and VEGF expression; in addition, thalidomide preserved fast-twitch skeletal muscle fibers and was associated with decreased expression of TNFalpha and TGFbeta1.
Collapse
Affiliation(s)
- Keng-Hao Liu
- Department of Surgery, Xiamen Chang Gung and Hospital Chang Gung Memorial Hospital, Chang Gung University, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
59
|
Abstract
Small-cell lung cancer is an aggressive form of lung cancer that, overall, remains the most common cause of cancer death in the US. Some advances have been made in the treatment of small-cell lung cancer using cytotoxic chemotherapeutic agents but no truly targeted therapies are available as of yet. At present, research is focused on finding therapies that can target the specific molecular mechanisms responsible for the survival, growth and metastasis of the tumor thereby improving responses to chemotherapy and minimizing toxicity. Several new agents, such as angiogenesis inhibitors and regulators of apoptosis, have reached clinical testing and multiple others are in preclinical trials. Some of these will be discussed in this review.
Collapse
Affiliation(s)
- Khaled Fernainy
- Emory University School of Medicine and Crawford Long Hospital, Atlanta, GA 30308, USA
| | | |
Collapse
|
60
|
Chen SC, Tsai HJ, Jan CM, Wang YH, Chen LT. Low dose of thalidomide can be effective in advanced hepatocellular carcinoma. J Gastroenterol Hepatol 2006; 21:1868-9. [PMID: 17074035 DOI: 10.1111/j.1440-1746.2006.04303.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
|
61
|
Abstract
AIM: To evaluate which patients with hepatocellular carcinoma (HCC) are most likely to respond to thalidomide treatment.
METHODS: From July 2002 to July 2004, patients with HCC who received thalidomide treatment, were enrolled. We extracted relevant data from the patients’ medical records, including history and type of hepatitis, comorbidity, serum α-fetoprotein (α-FP) level, volumetric changes in tumor, length of survival, and the dose, duration, side effects of thalidomide treatment. The tumor response was evaluated. On the basis of these data, the patients were divided into two groups: those with either partial response or stable disease (PR + SD group) and those with progressive disease (PD group).
RESULTS: Two of 42 (5%) patients had a partial tumor response after treatment with thalidomide, 200 mg/d, and 9 (21%) had stable disease. Patients in the PR + SD group all had cirrhosis. Comparing patients with and without cirrhosis, the former were more likely to respond to thalidomide therapy (PR + SD: 100% vs PD: 64.5%, P = 0.041 < 0.05). Thalidomide was significantly more likely to be effective in tumors smaller than 5 cm (PR + SD: 63.6% vs PD: 25.8%, P = 0.034 < 0.05). Compared with patients with progressive disease (PD), patients in the PR + SD group had a higher total dose of thalidomide (13 669.4 ± 8446.0 mg vs 22 022.7 ± 11 461.4 mg, P = 0.023 < 0.05) and a longer survival (181.0 ± 107.1 d vs 304.4 ± 167.1 d, P = 0.047 < 0.05). Patients with comorbid disease had a significantly greater incidence of adverse reactions than those without (93.8% vs 60.0%, P = 0.021 < 0.05). The average number of adverse reactions in each person with a comorbid condition was twice as high as in those without other diseases (2.2 ± 1.3 vs 1.1 ± 1.2; P = 0.022 < 0.05).
CONCLUSION: Thalidomide therapy is most likely to be effective in patients with early stage small HCC, especially in those with other underlying diseases. A low dose (200 mg/d) of thalidomide is recommended to continue the treatment long enough to make it more effective.
Collapse
Affiliation(s)
- Hsueh-Erh Chiou
- Pharmacy Department, Mackay Memorial Hospital, Taipei 10449, Taiwan, China
| | | | | | | |
Collapse
|
62
|
Abstract
Worldwide, hepatocellular carcinoma (HCC) is the fifth most common cancer and the third most common cause of cancer-related death. In the U.S., 18,510 new cancers of the liver and intrahepatic bile duct are expected in 2006, with an estimated 16,200 deaths. The incidence rates for HCC in the U.S. continued to rise steadily through 1998 and doubled during the period 1975-1995. Unresectable or metastatic HCC carries a poor prognosis, and systemic therapy with cytotoxic agents provides marginal benefit. A majority of HCC patients (>80%) presents with advanced or unresectable disease. Even for those with resected disease, the recurrence rate can be as high as 50% at 2 years. Because of the poor track record of systemic therapy in HCC, there has been a sense of nihilism for this disease in the oncology community for decades. However, with the arrival of newly developed molecularly targeted agents and the success of some of these agents in other traditionally challenging cancers, like renal cell carcinoma, there has recently been renewed interest in developing systemic therapy for HCC. This review attempts to concisely summarize the historical perspective and the current status of systemic therapy development in HCC.
Collapse
Affiliation(s)
- Andrew X Zhu
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts 02114, USA.
| |
Collapse
|
63
|
Shiah HS, Chao Y, Chen LT, Yao TJ, Huang JD, Chang JY, Chen PJ, Chuang TR, Chin YH, Whang-Peng J, Liu TW. Phase I and pharmacokinetic study of oral thalidomide in patients with advanced hepatocellular carcinoma. Cancer Chemother Pharmacol 2006; 58:654-64. [PMID: 16520988 DOI: 10.1007/s00280-006-0203-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2005] [Accepted: 01/30/2006] [Indexed: 10/24/2022]
Abstract
PURPOSE To evaluate the dose-limiting toxicities (DLT), maximum tolerated dose (MTD), and pharmacokinetics of thalidomide in patients with advanced hepatocellular carcinoma (HCC). METHODS Patients with advanced HCC who were not feasible for definitive local therapy were eligible. Patients were enrolled in a cohort of three to receive thalidomide twice daily for 1 week to determine the MTD. Intra-patient dose escalation was permitted. Pharmacokinetic studies were performed at the first dose level and repeated at the second dose level of each patient. RESULTS Fifteen patients were accrued at four dose levels with the starting dose range 100-400 mg/day. Two patients at 400 mg/day experienced DLT (grade 3 angioedema and dyspnea, respectively). The MTD of twice-daily schedule was determined as 300 mg/day. The mean steady-state maximal blood concentration and mean steady-state area under the curve had a trend toward positive correlation, but non-linear proportionate, to the daily dose of thalidomide. Pharmacokinetic parameters are comparable for patients of Child-Pugh's A and B. Apparent mild, transient drug-induced transaminitis was early onset, self-limited, which occurred in 30.7% of patients. Serum hepatitis B or C viral titers was largely not affected. CONCLUSION The absorption and elimination of thalidomide are not significantly different in HCC patients with compensated or decompensated hepatic dysfunction.
Collapse
Affiliation(s)
- Her-Shyong Shiah
- Division of Cancer Research, National Health Research Institutes, Ward 191 Veterans General Hospital, Taipei, and Department of Internal Medicine, Kaohsiung Medical University Hospital, Taiwan, ROC
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Kim DH, Choe YS, Jung KH, Lee KH, Choi Y, Kim BT. Synthesis and evaluation of 4-[18F]fluorothalidomide for the in vivo studies of angiogenesis. Nucl Med Biol 2006; 33:255-62. [PMID: 16546681 DOI: 10.1016/j.nucmedbio.2005.12.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2005] [Revised: 12/06/2005] [Accepted: 12/06/2005] [Indexed: 11/21/2022]
Abstract
In this study, we prepared 2-(2,6-dioxopiperidin-3-yl)-4-[(18)F]fluoroisoindole-1,3-dione (4-[(18)F]fluorothalidomide; [(18)F]1) for the in vivo studies of angiogenesis. Radiochemical synthesis of [(18)F]1 was carried out by labeling 4-trimethylammoniumthalidomide trifluoromethanesulfonate with nBu(4)N[(18)F]F in dimethyl sulfoxide (DMSO), followed by reverse-phase HPLC purification. Decay-corrected radiochemical yield of [(18)F]1 was 50-60%, with an effective specific activity of 42-120 GBq/micromol (end of synthesis). Incubation of the radioligand with human umbilical vein endothelial cells (HUVEC-C; American Type Culture Collection) showed a time-dependent increase in the uptake of the radioligand, and the uptake was inhibited by 8-11% in the presence of 10 microM thalidomide, indicating nonspecific binding of the radioligand. Positron emission tomography (PET) images of mice implanted with tumors in their right flanks revealed a marked accumulation of radioactivity in the livers, kidneys and bladders of the mice, and brain uptake appeared at approximately 40 min after injection. However, no radioactivity uptake was detected in the implanted tumor. Thin-layer chromatography (TLC), HPLC and LC-MS analyses of mouse liver microsomal metabolites of [(18)F]1 and 1 with or without nicotinamide adenine dinucleotide phosphate (NADPH) clearly revealed that the radioligand did not go through metabolic activation but underwent nonenzymatic hydrolysis at physiological pH. Therefore, these results would appear to indicate that [(18)F]1 may not be suitable for the in vivo studies of angiogenesis at least in mice, although it was reported that thalidomide and/or its hydrolysis products may be responsible for its activity in humans.
Collapse
Affiliation(s)
- Dong Hyun Kim
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 135-710, South Korea
| | | | | | | | | | | |
Collapse
|
65
|
Haffner MC, Berlato C, Doppler W. Exploiting our knowledge of NF-kappaB signaling for the treatment of mammary cancer. J Mammary Gland Biol Neoplasia 2006; 11:63-73. [PMID: 16900390 DOI: 10.1007/s10911-006-9013-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Nuclear Factor-kappaB (NF-kappaB) has been implicated in the lobuloalveolar development of the mammary gland. In breast cancer its activation has been linked to tumor progression via stimulation of cell proliferation, pro-survival, and angiogenesis pathways and metastasis. Whether NF-kappaB activation in the immune system influences mammary cancer remains unclear. In addition to the constitutive activation frequently found in mammary carcinoma tissue, radio- and chemotherapeutic agents used in the treatment of mammary cancer can lead to activation of NF-kappaB. This effect has been postulated to contribute to the development of resistance to these agents and suggests the use of NF-kappaB inhibitors as sensitizers for therapy. The review describes principle targets and drugs used to inhibit NF-kappaB function and discusses future perspectives in the use of these inhibitors for the treatment of mammary cancer.
Collapse
Affiliation(s)
- Michael C Haffner
- Division Medical Biochemistry, Biocenter, Innsbruck Medical University, Fritz-Pregl-Str. 3, A-6020 Innsbruck, Austria
| | | | | |
Collapse
|
66
|
Downs LS, Rogers LM, Yokoyama Y, Ramakrishnan S. Thalidomide and angiostatin inhibit tumor growth in a murine xenograft model of human cervical cancer. Gynecol Oncol 2005; 98:203-10. [PMID: 15975645 DOI: 10.1016/j.ygyno.2005.04.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2004] [Revised: 04/25/2005] [Accepted: 04/28/2005] [Indexed: 11/19/2022]
Abstract
OBJECTIVE To determine the impact of thalidomide and angiostatin on tumor growth, angiogenesis, and apoptosis in a xenograft model of cervical cancer. METHODS Human umbilical endothelial cells were treated with angiostatin or thalidomide and bFGF-induced proliferation was assessed with the MTT assay. Human cervical cancer cells (CaSki and SiHa) were injected into the flanks of nude mice. After tumors developed, mice were treated with angiostatin 20 mg/kg/day or thalidomide 200 mg/kg/day for 30 days. Fractional tumor growth was determined and immunohistochemical analysis of tumors was used to determine degree of angiogenesis. TUNEL assay was used to assess apoptosis. RESULTS Angiostatin inhibited endothelial cell proliferation by 50-60%. Thalidomide had no direct effect on endothelial cells. Angiostatin and thalidomide both inhibited tumor growth by about 55%. We found no additive or synergistic effect when the two agents were combined. Both agents inhibited angiogenesis and induced apoptosis when compared to tumors from control animals. CONCLUSIONS Angiostatin and thalidomide inhibit tumor growth, angiogenesis, and induce apoptosis in this xenograft model of cervical cancer.
Collapse
Affiliation(s)
- Levi S Downs
- The University of Minnesota Medical School, Department of Obstetrics, Gynecology and Women's Health, Division of Gynecologic Oncology, 420 Delaware Street SE, MMC 395, Minneapolis, MN 55455, USA.
| | | | | | | |
Collapse
|
67
|
Reiriz AB, Richter MF, Fernandes S, Cancela AI, Costa TD, Di Leone LP, Schwartsmann G. Phase II study of thalidomide in patients with metastatic malignant melanoma. Melanoma Res 2005; 14:527-31. [PMID: 15577325 DOI: 10.1097/00008390-200412000-00014] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Thalidomide has anti-angiogenic and immunomodulatory activity, exhibiting antitumour effects in patients with multiple myeloma and, more rarely, in several other solid tumours. We evaluated the single-agent antitumour activity and toxicity profile of thalidomide in patients with metastatic malignant melanoma, as well as its plasma pharmacokinetics and pharmacodynamic effects [vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (b-FGF) levels]. A two-stage Gehan method was used with a stopping rule after 14 consecutive non-responding patients. Thalidomide was given orally at a daily dose of 200 mg/day, which was then escalated every 2 weeks by 200 mg/day as tolerated to a maximum of 800 mg/day. Patients were evaluated every 8 weeks for response using the World Health Organization (WHO)-27 criteria. Fourteen patients were enrolled and no objective responses were observed, with one stable disease and one mixed response. The dose-limiting toxicities were constipation, dizziness and somnolence. Other toxicities were oedema, neuropathy, dry skin, dry mouth, tremor and fatigue. The plasma pharmacokinetics of thalidomide were comparable with those of previous studies in normal volunteers and in patients with advanced prostate cancer. Serum levels of b-FGF and VEGF did not change significantly following drug administration. In conclusion, thalidomide showed poor activity, but acceptable toxicity, in patients with metastatic melanoma. Future studies should explore this agent in combination with other biological agents or cytotoxic agents, such as temozolomide.
Collapse
Affiliation(s)
- André B Reiriz
- South American Office for Anticancer Drug Development, Porto Alegre, RS, Brazil.
| | | | | | | | | | | | | |
Collapse
|
68
|
Staehler M, Rohrmann K, Bachmann A, Zaak D, Stief CG, Siebels M. Therapeutic approaches in metastatic renal cell carcinoma. BJU Int 2005; 95:1153-61. [PMID: 15877725 DOI: 10.1111/j.1464-410x.2005.05537.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Michael Staehler
- Department of Urology, Klinikum Grosshadern, Ludwig Maximilians University Munich, Germany
| | | | | | | | | | | |
Collapse
|
69
|
Laviano A, Meguid MM, Inui A, Muscaritoli M, Rossi-Fanelli F. Therapy Insight: cancer anorexia–cachexia syndrome—when all you can eat is yourself. ACTA ACUST UNITED AC 2005; 2:158-65. [PMID: 16264909 DOI: 10.1038/ncponc0112] [Citation(s) in RCA: 229] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2004] [Accepted: 01/28/2005] [Indexed: 12/20/2022]
Abstract
Tumor growth is associated with profound metabolic and neurochemical alterations, which can lead to the onset of anorexia-cachexia syndrome. Anorexia is defined as the loss of the desire to eat, while cachexia results from progressive wasting of skeletal muscle mass--and to a lesser extent adipose tissue--occurring even before weight loss becomes apparent. Cancer anorexia-cachexia syndrome is highly prevalent among cancer patients, has a large impact on morbidity and mortality, and impinges on patient quality of life. However, its clinical relevance is frequently overlooked, and treatments are usually only attempted during advanced stages of the disease. The pathogenic mechanisms of cachexia and anorexia are multifactorial, but cytokines and tumor-derived factors have a significant role, thereby representing a suitable therapeutic target. Energy expenditure in anorexia is frequently increased while energy intake is decreased, which further exacerbates the progressive deterioration of nutritional status. The optimal therapeutic approach to anorectic-cachectic cancer patients should be based on both changes in dietary habits, achieved via nutritional counseling; and drug therapy, aimed at interfering with cytokine expression or activity. Our improved understanding of the influence a tumor has on the host's metabolism is advancing new therapeutic approaches, which are likely to result in better preservation of nutritional status if started concurrently with specific antineoplastic treatment.
Collapse
|
70
|
Sharma R, Tobin P, Clarke SJ. Management of chemotherapy-induced nausea, vomiting, oral mucositis, and diarrhoea. Lancet Oncol 2005; 6:93-102. [PMID: 15683818 DOI: 10.1016/s1470-2045(05)01735-3] [Citation(s) in RCA: 132] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The past 10 years have seen substantial advances in molecularly targeted therapies for treatment of patients with cancer; however, chemotherapy will continue to be used. Therefore, the toxic effects of chemotherapy must be readily managed-especially nausea, vomiting, mucositis, and diarrhoea. For moderately to highly emetogenic chemotherapy, standard prophylactic treatment is an antagonist for 5-hydroxytryptamine 3 receptors (5-HT3R) combined with dexamethasone for the acute phase, and dexamethasone with another agent for prevention of the delayed phase. Palonoestron (a 5-HT3R antagonist) and aprepitant (an antagonist for the protachykinin 1 receptor) have been introduced for the prevention of emesis. Other agents such as cannabinoids, gabapentin, and olanzapine might also be effective. There is no standard prophylactic regimen for chemotherapy-induced mucositis. The most common treatment is optimum care of the mouth by use of mouthwashes. Keratinocyte growth factor, molgromastim, and transforming growth factor beta3 may also reduce chemotherapy-induced mucositis. Severe diarrhoea is another potentially fatal complication of chemotherapy and is most common in patients treated with irinotecan. Several interventions have been assessed for prevention and treatment of diarrhoea such as high-dose loperamide, non-absorbable antibiotics, budesonide, thalidomide, and fish oils, but only loperamide is used routinely. Symptom management has become a focus of clinical research, and development of personalised medicine should identify patients at increased risk of toxic effects because of molecular or biochemical factors, thus leading to changes in dose, early intervention, or use of alternative therapies.
Collapse
Affiliation(s)
- Rohini Sharma
- Sydney Cancer Centre, Camperdown, New South Wales, Australia
| | | | | |
Collapse
|
71
|
Abstract
PURPOSE OF REVIEW Despite advances in surgery, radiation therapy, and chemotherapy, malignant gliomas continue to be associated with a poor prognosis. Even the most intensive combinations of radiotherapy and chemotherapy are not curative. In recent years our understanding of how tumor cells overcome cell cycle control, evade programmed cell death, induce blood vessel formation, and escape immune regulation has increased substantially. Significant efforts are directed towards the development of novel experimental therapies to target these molecular and biological mechanisms that lead to the development and growth of brain tumors. This review summarizes the most recent developments in non-cytotoxic therapy for malignant gliomas, such as targeted molecular drugs, inhibitors of angiogenesis and intratumoral therapy. RECENT FINDINGS The first generation of studies using these novel therapies is nearing completion. In general, most of these treatments are well tolerated, but single-agent activity is modest. There is significant interest in combining these therapies with each other and with conventional cytotoxic therapies such as radiation therapy and chemotherapy. SUMMARY These new therapeutic approaches for malignant gliomas are showing modest activity. As we learn to use these agents more effectively, and as an increasing number of new and potentially promising agents are developed, it is likely that therapies for malignant gliomas will improve over the next few years.
Collapse
Affiliation(s)
- Jan Drappatz
- Center for Neuro-Oncology, Dana Farber Cancer Institute, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | | |
Collapse
|
72
|
Highlights From: 40th Annual Meeting of the American Society of Clinical Oncology; June 2004 New Orleans, Louisiana. ACTA ACUST UNITED AC 2004; 5:74-80. [PMID: 15453920 DOI: 10.1016/s1526-9655(11)70059-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|