1
|
Negahdary M. Aptamers in nanostructure-based electrochemical biosensors for cardiac biomarkers and cancer biomarkers: A review. Biosens Bioelectron 2020; 152:112018. [PMID: 32056737 DOI: 10.1016/j.bios.2020.112018] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 12/16/2019] [Accepted: 01/09/2020] [Indexed: 01/01/2023]
Abstract
Heart disease (especially myocardial infarction (MI)) and cancer are major causes of death. Recently, aptasensors with the applying of different nanostructures have been able to provide new windows for the early and inexpensive detection of these deadly diseases. Early, inexpensive, and accurate diagnosis by portable devices, especially aptasensors can increase the likelihood of survival as well as significantly reduce the cost of treatment. In this review, recent studies based on the designed aptasensors for the diagnosis of these diseases were collected, ordered, and reviewed. The biomarkers for the diagnosis of each disease were discussed separately. The primary constituent elements of these aptasensors including, analyte, aptamer sequence, type of nanostructure, diagnostic technique, analyte detection range, and limit of detection (LOD), were evaluated and compared.
Collapse
Affiliation(s)
- Masoud Negahdary
- Yazd Cardiovascular Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
2
|
Abstract
The kinase inhibitors sorafinib and sunitinib have demonstrated significant activity in renal cell carcinoma (RCC), and are now approved by the FDA for use in advanced disease. There still remains a need for novel therapies. Our group were the first to demonstrate activity of thalidomide in RCC, believed to be in part related to the modulation of tumor necrosis factor (TNF-a), a cytokine secreted by RCC with a number of tumor promoting properties. We subsequently conducted a phase II trial of the TNF-a monoclonal antibody infliximab in patients with previously treated advanced RCC. The drug was well tolerated. The response rate was 16% and stability was achieved in a further 16% of patients. Anti-TNF-a therapy may represent an important approach in the treatment of this disease.
Collapse
Affiliation(s)
- Nick Maisey
- Department of Academic Oncology, Guys Hospital, London, United Kingdom.
| |
Collapse
|
3
|
Harshman LC, Srinivas S. The bevacizumab experience in advanced renal cell carcinoma. Onco Targets Ther 2010; 3:179-89. [PMID: 21049084 PMCID: PMC2962304 DOI: 10.2147/ott.s8157] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Indexed: 11/23/2022] Open
Abstract
Bevacizumab in combination with interferon alfa is now approved for treatment-naïve advanced renal cell carcinoma (RCC) in both the US and Europe. Its objective response rates of 30% and progression-free survival rates of 9–10 months are comparable to the other approved first-line multityrosine kinase inhibitors, sunitinib and pazopanib. Its advantages include a different toxicity profile and assurance of administration compliance given its intravenous formulation. Enthusiasm for its use is blunted by the increased costs, the potential infusion-related reactions, the associated interferon-related toxicities, and the inconvenience of its nonoral formulation. Further study is warranted to assess its efficacy both as a single agent and in combination with the targeted agents and other immunotherapies. With multiple agents now available for the treatment of advanced RCC, identification of patient and tumor-specific biomarkers to inform our choice of first-line therapy and the proper sequence of subsequent therapies is imperative.
Collapse
Affiliation(s)
- Lauren C Harshman
- Division of Oncology, Stanford University School of Medicine, Stanford, California, USA
| | | |
Collapse
|
4
|
TAN TH, PRANAVAN G, HAXHIMOLLA HZ, YIP D. New systemic treatment options for metastatic renal-cell carcinoma in the era of targeted therapies. Asia Pac J Clin Oncol 2010; 6:5-18. [DOI: 10.1111/j.1743-7563.2010.01277.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
5
|
Greenstein RJ, Su L, Brown ST. On the effect of thalidomide on Mycobacterium avium subspecies paratuberculosis in culture. Int J Infect Dis 2009; 13:e254-63. [PMID: 19303801 DOI: 10.1016/j.ijid.2008.10.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2008] [Accepted: 10/24/2008] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Without known mechanisms of action, thalidomide is used to treat a variety of non-malignant 'idiopathic' diseases. There is increasing concern that Mycobacterium avium subspecies paratuberculosis (MAP) may be zoonotic. Recently, methotrexate, azathioprine, 6-mercaptopurine (6-MP), 5-aminosalicylic acid (5-ASA), cyclosporine A, rapamycin, and tacrolimus have been shown to inhibit MAP growth in culture, indicating that, unknowingly, MAP infections may have been treated for decades. We herein test the hypothesis that thalidomide may inhibit MAP growth. METHODS Using the radiometric 14CO2 (Bactec) system we quantified growth kinetics of thalidomide (+/-), (+), and (-) and two components for thalidomide, phthalimide and 1-hydroxypiperidine-2,6-dione (HPD). We studied four MAP strains (three human isolates, 'Ben', 'Dominic', and UCF-4, and a bovine MAP isolate 19698) and three mycobacterial controls (Mycobacterium avium and bacillus Calmette-Guérin (BCG)). Growth was quantified as growth index (GI) and inhibition as percent decrease in cumulative GI (%-DeltacGI). RESULTS Phthalimide had no dose-dependent inhibition on any strain. Neither thalidomide nor HPD inhibited M. avium or BCG. MAP inhibition varied; at 64 microg/ml, amongst human isolates, Dominic was most susceptible: thalidomide (+)=58%-DeltacGI and HPD=46%-DeltacGI. UCF-4 was next: thalidomide (-)=37%-DeltacGI and HPD=40%-DeltacGI. Ben was least susceptible: HPD=24%-DeltacGI. CONCLUSIONS We have shown, in culture, the heretofore-undescribed inhibition of MAP growth by thalidomide and its enantiomers. Phthalimide was found to have no anti-MAP activity, whereas HPD was found to inhibit MAP growth. These data are compatible with the hypothesis that thalidomide, like other 'anti-inflammatories' and 'immunomodulators' may act, in part, as an anti-MAP antibiotic.
Collapse
|
6
|
The Combination of Thalidomide and Capecitabine in Metastatic Renal Cell Carcinoma—Is Not the Answer. Am J Clin Oncol 2008; 31:417-23. [DOI: 10.1097/coc.0b013e318168ef47] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
7
|
Phase II trial of lenalidomide in patients with metastatic renal cell carcinoma. Invest New Drugs 2007; 26:273-6. [DOI: 10.1007/s10637-007-9107-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2007] [Accepted: 12/06/2007] [Indexed: 10/22/2022]
|
8
|
Renal Cell Cancer. Oncology 2007. [DOI: 10.1007/0-387-31056-8_46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
9
|
Harrison ML, Obermueller E, Maisey NR, Hoare S, Edmonds K, Li NF, Chao D, Hall K, Lee C, Timotheadou E, Charles K, Ahern R, King DM, Eisen T, Corringham R, DeWitte M, Balkwill F, Gore M. Tumor necrosis factor alpha as a new target for renal cell carcinoma: two sequential phase II trials of infliximab at standard and high dose. J Clin Oncol 2007; 25:4542-9. [PMID: 17925549 DOI: 10.1200/jco.2007.11.2136] [Citation(s) in RCA: 189] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
PURPOSE Tumor necrosis factor alpha (TNF-alpha) may play a role in renal cell carcinoma (RCC). We performed two sequential phase II studies of infliximab, an anti-TNF-alpha monoclonal antibody, in patients with immunotherapy-resistant or refractory RCC. PATIENTS AND METHODS Patients progressing after cytokine therapy were treated with intravenous infliximab as follows: study 1 (19 patients), 5 mg/kg at weeks 0, 2, and 6, and then every 8 weeks; study 2 (18 patients), 10 mg/kg at weeks 0, 2, and 6, and then every 4 weeks. Treatment continued until disease progression (PD). Response was assessed according to Response Evaluation Criteria in Solid Tumors. Plasma levels of TNF-alpha, CCL2, and interleukin-6 (IL-6) were measured before and during treatment. RESULTS TNF-alpha and its receptors were detected in malignant cells in RCC biopsies. In study 1, three patients (16%) achieved partial response (PR) and three patients (16%) achieved stable disease (SD). Median duration of response (PR + SD) was 7.7 months (range, 5.0 to 40.5+ months). In study 2, 11 patients (61%) achieved SD. Median duration of response was 6.2 months (range, 3.5 to 24+ months). One patient developed grade 3 hypersensitivity and another died as a result of pulmonary infection/sepsis. Enzyme-linked immunosorbent assay analysis of plasma revealed that higher levels of TNF-alpha at baseline and higher levels of CCL2 during treatment were associated with PD. There were also correlations between higher levels of TNF-alpha, IL-6, and CCL2 and poor survival (< 12 months). CONCLUSION This is the first direct clinical evidence suggesting that TNF-alpha may be a therapeutic target in RCC. Plasma levels of TNF-alpha, IL-6, and CCL2 may have predictive and prognostic significance.
Collapse
|
10
|
Amato RJ, Khan M. A phase I clinical trial of low-dose interferon-alpha-2A, thalidomide plus gemcitabine and capecitabine for patients with progressive metastatic renal cell carcinoma. Cancer Chemother Pharmacol 2007; 61:1069-73. [PMID: 17701037 DOI: 10.1007/s00280-007-0568-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2007] [Accepted: 07/26/2007] [Indexed: 11/24/2022]
Abstract
BACKGROUND We have conducted a phase I trial to determine the maximum tolerated dose of gemcitabine in combination with interferon, thalidomide and capecitabine. METHODS Patients received oral capecitabine 1,000 mg/m(2 )per day, divided in 2 daily doses, 2 weeks on, 1 week off; subcutaneous interferon-alpha 1 mIU twice a day without an interruption; daily oral thalidomide 200 mg/day for the first 7 days, then escalated to 400 mg/day without an interruption. Gemcitabine was given by intravenous administration over 30 min on day 1, week 1 and day 8, week 2. Initial dose level of gemcitabine was 400 mg/m(2). The dose of gemcitabine was the phase I variable. One cycle was 3 weeks. RESULTS AND DISCUSSION We treated 12 patients, 6 patients were entered at a dose level of 0 (gemcitabine 400 mg/m(2)) and 6 patients entered at a dose level-1 (gemcitabine 200 mg/m(2)). Eight of 12 patients completed at least 12 weeks of therapy. Three partial responses and two stable disease were observed. The remaining patients had progressive disease. Non-hematologic toxicity was either grade 1 or 2. Hematologic toxicity at dose level 0 consisted of 3 patients with grade 3/4 neutropenia, and 1 patient with grade 3 thrombocytopenia. At dose level-1 grade 1/2 neutropenia was observed. CONCLUSIONS The completion of our phase I experience determined our maximum tolerated dose to be dose level-1. The phase II trial is currently being proposed for patients with rapidly growing clear cell, other histologies that may contain sarcomatoid elements or collecting duct tumor.
Collapse
Affiliation(s)
- Robert J Amato
- The Methodist Hospital Research Institute, Genitourinary Oncology Program, 6560 Fannin Street, Suite 2050, Houston, TX 77030, USA.
| | | |
Collapse
|
11
|
Abstract
Multidrug resistance is a major obstacle to successful cancer treatment. One mechanism by which cells can become resistant to chemotherapy is the expression of ABC transporters that use the energy of ATP hydrolysis to transport a wide variety of substrates across the cell membrane. There are three human ABC transporters primarily associated with the multidrug resistance phenomenon, namely Pgp, MRP1, and ABCG2. All three have broad and, to a certain extent, overlapping substrate specificities, transporting the major drugs currently used in cancer chemotherapy. ABCG2 is the most recently described of the three major multidrug-resistance pumps, and its substrates include mitoxantrone, topotecan, irinotecan, flavopiridol, and methotrexate. Despite several studies reporting ABCG2 expression in normal and malignant tissues, no trials have thus far addressed the role of ABCG2 in clinical drug resistance. This gives us an opportunity to critically review the disappointing results of past clinical trials targeting Pgp and to propose strategies for ABCG2. We need to know in which tumor types ABCG2 contributes to the resistance phenotype. We also need to develop standardized assays to detect ABCG2 expression in vivo and to carefully select the chemotherapeutic agents and clinical trial designs. This review focuses on our current knowledge about normal tissue distribution, tumor expression profiles, and substrates and inhibitors of ABCG2, together with lessons learned from clinical trials with Pgp inhibitors. Implications of SNPs in the ABCG2 gene affecting the pharmacokinetics of substrate drugs, including many non-chemotherapy agents and ABCG2 expression in the SP population of stem cells are also discussed.
Collapse
Affiliation(s)
- Robert W Robey
- Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
12
|
Choueiri TK, Dreicer R, Rini BI, Elson P, Garcia JA, Thakkar SG, Baz RC, Mekhail TM, Jinks HA, Bukowski RM. Phase II study of lenalidomide in patients with metastatic renal cell carcinoma. Cancer 2006; 107:2609-16. [PMID: 17075879 DOI: 10.1002/cncr.22290] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Lenalidomide (LEN) is a structural and functional analogue of thalidomide that has demonstrated enhanced immunomodulatory properties and a more favorable toxicity profile. A Phase II, open-label study of LEN in patients with metastatic renal cell carcinoma (RCC) was conducted to determine its safety and clinical activity. METHODS Patients with metastatic RCC received LEN orally at a dose of 25 mg daily for the first 21 days of a 28-day cycle. The primary endpoint was the objective response rate. Time to treatment failure, safety, and survival were secondary endpoints. RESULTS In total, 28 patients participated in the trial and were included in the current analysis. Three of 28 patients (11%) demonstrated partial responses and continued to be progression-free for >15 months. Eleven patients (39%) had stable disease that lasted >3 months, including 8 patients who had tumor shrinkage. In total, 6 patients (21%) remained on the trial, and 5 additional patients continued to be followed for survival. The median follow-up for those 11 patients was 13.5 months (range, 8.3-17.0 months). The median survival had not been reached at the time of the current report. Serious adverse events included fatigue (11%), skin toxicity (11%), and neutropenia (36%). CONCLUSIONS LEN demonstrated an antitumor effect in metastatic RCC, as evidenced by durable partial responses. LEN toxicities were manageable. Further studies will be required to assess the overall activity of LEN in patients with metastatic RCC.
Collapse
Affiliation(s)
- Toni K Choueiri
- Cleveland Clinic Taussig Cancer Center, Cleveland, Ohio, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Amato RJ, Rawat A. Interferon-alpha plus capecitabine and thalidomide in patients with metastatic renal cell carcinoma: a pilot study. Invest New Drugs 2006; 24:171-5. [PMID: 16086096 DOI: 10.1007/s10637-005-2938-5] [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] [Indexed: 10/25/2022]
Abstract
PURPOSE To assess the activity and toxicity of interferon-alpha (IFN-alpha), capecitabine, and thalidomide in patients with metastatic renal cell carcinoma (MRCC). PATIENTS AND METHODS Twenty-seven patients were enrolled in a pilot study to receive oral capecitabine 1,900 mg/m2/day in 2 daily doses, 2 weeks on, l week off; daily subcutaneous IFN-alpha 1 mIU without interruption; and daily oral thalidomide 200 mg/day for the first seven days, then escalated to 400 mg/day without interruption. Dosages were reduced for toxicity as necessary. RESULTS Two patients discontinued treatment during the first week of the study, leaving 25 patients evaluable. There were 5 (20%) partial responses (PRs), 1 (4%) minor response (MR), 6 (24%) cases of stable disease (SD) > or = 6 months, and 13 (52%) cases of progressive disease (PD). The interval from first response to disease progression varied from 0-23 months: 17 patients progressed in 0-6 months; 4 progressed in 7-12 months; and 4 progressed in 12-24 months. Median survival was > 22 months, 14 months, and 1 month, respectively, for patients with PR, SD, and PD. Grade 3/4 toxicities consisted of hand-foot syndrome, neuropathy, fatigue, anemia, and deep venous thrombosis were common. CONCLUSION This study demonstrates antitumor activity of combination IFN-alpha/capecitabine/thalidomide in MRCC. The 20% PR rate was notable, as the patient population had advanced disease and inferior performance status. Treatment was generally well tolerated, and further research is warranted to explore the efficacy of this combination for treating MRCC.
Collapse
Affiliation(s)
- Robert J Amato
- Genitourinary Oncology Program, The Methodist Hospital, Research Institute, Houston, TX 77030, USA.
| | | |
Collapse
|
14
|
Lee CP, Patel PM, Selby PJ, Hancock BW, Mak I, Pyle L, James MG, Beirne DA, Steeds S, A'Hern R, Gore ME, Eisen T. Randomized Phase II Study Comparing Thalidomide With Medroxyprogesterone Acetate in Patients With Metastatic Renal Cell Carcinoma. J Clin Oncol 2006; 24:898-903. [PMID: 16484699 DOI: 10.1200/jco.2005.03.7309] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PurposeTo investigate escalating doses of thalidomide compared with medroxyprogesterone in patients with metastatic renal cell carcinoma (RCC), who had either progressed after first-line immunotherapy or who were not suitable for immunotherapy.Patients and MethodsThalidomide was started at 100 mg/d orally (PO) and escalated by 100 mg/d every 2 weeks to the maximum dose of 400 mg/d. Medroxyprogesterone was given at a fixed dose of 300 mg PO daily.ResultsSixty patients were entered (thalidomide:medroxyprogesterone = 29:31; median age, 59 [thalidomide], 60 [medroxyprogesterone]; No. of patients assessable for response, 22 [thalidomide], 26 [medroxyprogesterone]). In the thalidomide arm, there was no objective response seen. The best response was SD in three patients lasting 5+, 6+, and 12 months, respectively. All patients in the medroxyprogesterone arm progressed. There was no difference in overall survival between the two arms; median survival in the thalidomide arm was 8.2 months compared with 4.8 months in the medroxyprogesterone arm (P = .62). Hazard ratio was 0.88 (95% CI, 0.67 to 1.94). Median duration of treatment was 73 days (range, 14 to 364 days) in the thalidomide arm, and 84 days (range, 7 to 175 days) in the medroxyprogesterone arm. The high incidence of toxicity in the thalidomide arm, mainly somnolence, constipation, fatigue and paraesthesia, meant that only 30.8% of patients were able to tolerate the maximum dose of 400 mg/d of treatment.ConclusionThalidomide is not superior to medroxyprogesterone acetate in patients with metastatic RCC. Its risk/benefit ratio does not favor its use in this patient population.
Collapse
Affiliation(s)
- Chooi P Lee
- Department of Medicine, Royal Marsden Hospital, London, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Abstract
Renal cell carcinoma (RCC) still represents a therapeutic challenge when patients have advanced or metastatic disease. Treatment using IL-2 and IFN-alpha continues to be the standard of care in patients who are able to tolerate such regimens. Targeted therapy may become the first-line treatment for patients resistant or intolerant to cytokines as new emerging drugs continue to be investigated. Understanding the genetic abnormalities related to the development of RCC (e.g., VHL gene abnormalities) and identifying molecular targets (e.g., epidermal growth factor, vascular endothelial growth factor and carbonic anhydrase IX) are playing a major role in the emergence of these novel agents for the treatment of this malignancy. Overall, these drugs are better tolerated and more acceptable to use by patients than the traditional cytokine-based regimens. The use of oral drugs to treat various malignancies including RCC seems to be the new paradigm of the future. Further understanding of their mechanisms of action and confirmation of their benefits on the clinical outcome is needed.
Collapse
Affiliation(s)
- Philip E Shaheen
- Cleveland Clinic Foundation, Experimental Therapeutics Program, Taussig Cancer Center, R33, Cleveland, OH 44195, USA
| | | |
Collapse
|
16
|
Srinivas S, Guardino AE. A lower dose of thalidomide is better than a high dose in metastatic renal cell carcinoma. BJU Int 2005; 96:536-9. [PMID: 16104906 DOI: 10.1111/j.1464-410x.2005.05680.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To conduct a dose-finding trial using a single low dose and dose escalation of a higher dose of thalidomide in patients with metastatic renal cell carcinoma (RCC), and to evaluate the antineoplastic effectiveness of thalidomide as an anti-angiogenic agent on RCC. PATIENTS AND METHODS The 14 patients enrolled in the study had progressive measurable metastatic RCC and consented to participate. Patients were randomized to either a fixed low dose of 200 mg of thalidomide or to a high dose of 800 mg that was increased to a maximum dose of 1200 mg daily. Patients were evaluated for response after 8 weeks of therapy. RESULTS Stable disease was achieved in six patients and was seen in both the low-dose and high-dose thalidomide groups. The median overall survival was 9 months. The low-dose thalidomide regimen was better tolerated and patients survived longer than those on the high-dose regimen (16 vs 6 months, P = 0.04) CONCLUSION The use of low-dose thalidomide in patients with metastatic RCC was well tolerated and they survived for longer than those on the high-dose regimen.
Collapse
Affiliation(s)
- Sandy Srinivas
- Division of Oncology, Stanford Medical Center, Stanford, CA, USA.
| | | |
Collapse
|
17
|
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
|
18
|
D'Hondt V, Gil T, Lalami Y, Piccart M, Awada A. Will the dark sky over advanced renal cell carcinoma soon become brighter? Eur J Cancer 2005; 41:1246-53. [PMID: 15939260 DOI: 10.1016/j.ejca.2004.11.025] [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: 11/17/2004] [Accepted: 11/21/2004] [Indexed: 01/02/2023]
Abstract
Until recently, immunotherapy has been the most efficient treatment for advanced renal cell carcinoma, but clinical results are largely unsatisfactory. More promising agents are being developed as a result of an improved understanding of the biology of the disease. Several agents that target known biological abnormalities of the disease are now being tested in the clinic. This review describes the encouraging clinical results obtained to date with these new drugs or combinations of drugs.
Collapse
Affiliation(s)
- V D'Hondt
- Clinic of Medical Oncology, Institut Jules Bordet, Bd de Waterloo 125, Brussels 1000, Belgium
| | | | | | | | | |
Collapse
|
19
|
Abstract
Thalidomide, an oral agent with antiangiogenic and immunomodulatory properties, is being investigated extensively in the management of advanced cancer. Multiple studies with large numbers of patients have confirmed that this drug has significant activity in multiple myeloma. Some patients with myelofibrosis or myeodysplatic syndromes may reduce their need for transfusions after thalidomide treatment. The activity of thalidomide in solid tumors is less prominent. Studies in Kaposi's sarcoma, malignant melanoma, renal cell carcinoma and prostate cancer appear more promising especially when thalidomide is combined with biological agents or with chemotherapy. Limited activity was demonstrated in patients with glioma, while thalidomide appears to be inactive in patients with head and neck cancer, breast or ovarian cancer.
Collapse
|
20
|
Rini BI, Small EJ. Biology and clinical development of vascular endothelial growth factor-targeted therapy in renal cell carcinoma. J Clin Oncol 2004; 23:1028-43. [PMID: 15534359 DOI: 10.1200/jco.2005.01.186] [Citation(s) in RCA: 289] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
PURPOSE To review the biology of renal cell carcinoma (RCC) leading to vascular endothelial growth factor (VEGF) overexpression and the clinical results of VEGF blockade in metastatic RCC. METHODS A review of relevant published literature regarding VEGF, von Hippel-Lindau (VHL) gene inactivation and VEGF overexpression in RCC was performed. Further, a review of the mechanism, toxicity, and clinical development of VEGF-targeted therapy in metastatic RCC was undertaken. RESULTS VEGF is the major proangiogenic protein that exerts a biologic effect through interaction with cellular receptors. The majority of sporadic clear-cell RCC tumors are characterized by VHL tumor suppressor gene inactivation. The resulting VHL gene silencing leads to VEGF overexpression. An antibody to VEGF (bevacizumab) has demonstrated a significant prolongation of time to disease progression compared with placebo in patients with metastatic RCC. Small molecules with inhibitory effects against the VEGF receptor have undergone initial clinical testing in metastatic RCC with substantial objective response rates. CONCLUSION Therapeutic targeting of VEGF in RCC has strong biologic rationale and preliminary clinical efficacy. Further investigation will determine the optimal timing, sequence, and utility of these agents in RCC.
Collapse
Affiliation(s)
- Brian I Rini
- University of California San Francisco Comprehensive Cancer Center, CA, USA.
| | | |
Collapse
|
21
|
Ebbinghaus SW, Gordon MS. Renal cell carcinoma: rationale and development of therapeutic inhibitors of angiogenesis. Hematol Oncol Clin North Am 2004; 18:1143-59, ix-x. [PMID: 15474339 DOI: 10.1016/j.hoc.2004.06.003] [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] [Indexed: 12/27/2022]
Abstract
Inhibition of tumor angiogenesis is a promising therapeutic approach to treat cancer; translation of this concept into clinical practice requires an understanding of the molecular events that are responsible for the development of tumor vasculature. Renal cell carcinoma is characterized by the frequent loss of the von Hippel-Lindau tumor suppressor gene which results in the loss of one of the critical mechanisms for regulating the level of hypoxia inducible factor 1 and leads to the overproduction of vascular endothelial growth factor (VEGF) by the tumor cell. Therapeutic strategies to inhibit the function of these important pathways have been effective in preventing tumor angiogenesis in preclinical models of kidney cancer, and more recently, in the clinical setting. Strategies to treat renal cell carcinoma with agents that are designed to prevent angiogenesis have included interruption of the VEGF signaling pathway, mimics of endogenous angiogenesis inhibitors, prevention of destruction of the basement membrane, and direct inhibition of endothelial cells by a variety of agents with complex, novel, or undetermined mechanisms. Recent clinical studies of bevacizumab, the first anti-VEGF agent to be marketed for the treatment of cancer, have provided proof for the concept that these strategies can lead to tangible benefits for patients who have advanced renal cell carcinoma and likely will be applicable broadly to the treatment of cancer.
Collapse
Affiliation(s)
- Scot W Ebbinghaus
- Division of Hematology-Oncology, University of Arizona College of Medicine, Arizona Cancer Center, 1515 North Campbell Avenue, Tucson, AZ 85724-5024, USA.
| | | |
Collapse
|
22
|
Abstract
Renal cell cancer remains a disease for which highly effective therapy for the majority of patients with metastatic disease is lacking. The biology of clear cell carcinomas and their association with mutations of the von Hippel-Lindau gene and its resultant increased expression of vascular endothelial growth factor (VEGF) make angiogenesis a potentially pathophysiologic mechanism for tumor development. As a result, the use of antiangiogenic therapy is an intriguing concept for the treatment of renal cell cancer. Various agents, aside from the inhibitors of VEGF, have been studied, including thalidomide, low-dose interferon, and novel antiangiogenic agents such as the thrombospondin-1 mimetics. Use of these agents has been associated with some degree of objective response or prolonged stabilization of disease, and their true value needs to be assessed in ongoing prospective studies. Combinations of antiangiogenic agents either with other similarly acting drugs or as a component of a "cocktail" with other noncytotoxic therapies should be explored in this patient population.
Collapse
Affiliation(s)
- Michael S Gordon
- Arizona Cancer Center-Greater Phoenix Area, University of Arizona College of Medicine, Scottsdale, Arizona 85258, USA.
| |
Collapse
|
23
|
Affiliation(s)
- S Vincent Rajkumar
- Division of Hematology, Mayo Clinic College of Medicine, Rochester, Minn 55905, USA.
| |
Collapse
|
24
|
Elaraj DM, White DE, Steinberg SM, Haworth L, Rosenberg SA, Yang JC. A pilot study of antiangiogenic therapy with bevacizumab and thalidomide in patients with metastatic renal cell carcinoma. J Immunother 2004; 27:259-64. [PMID: 15235386 PMCID: PMC2275325 DOI: 10.1097/00002371-200407000-00001] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The use of antiangiogenic agents represents a promising strategy for the treatment of patients with metastatic renal cell carcinoma. Objective responses to single-agent thalidomide have been described, and a randomized study showed that bevacizumab (a neutralizing antibody against vascular endothelial growth factor) delayed time to progression of metastatic renal cancer. A pilot study combining these two agents was performed. Sequential cohorts of 10 and 12 patients (crossing over from placebo therapy in the aforementioned randomized bevacizuamab trial) were treated with low-dose bevacizumab alone or bevacizumab plus the maximum tolerated dose of thalidomide as determined by intrapatient escalation. Toxicity, objective responses, and time to progression were the endpoints of this study. Patients tolerated thalidomide and bevacizumab well, with more than 50% of patients escalating to at least 500 mg/d thalidomide. Grades 1 and 2 sensory neuropathy limited thalidomide dose escalation in 3 of 12 patients. The incidence of grades 3 and 4 toxicity was not different between patients treated with bevacizumab alone versus bevacizumab plus thalidomide. There were no objective responses and no difference in progression-free survival between the groups (2.4 months for bevacizumab alone, 3.0 months for bevacizumab plus thalidomide). Combination antiangiogenic therapy with bevacizumab plus thalidomide in patients with renal cell carcinoma is associated with similar toxicity and progression-free survival compared with bevacizumab alone. This study illustrates a clinical trial design for rapidly testing the feasibility and safety of combining antiangiogenic agents, an approach that will be necessary for rapidly evaluating the many potential combinations of antiangiogenic agents.
Collapse
Affiliation(s)
- Dina M. Elaraj
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, U.S.A
| | - Donald E. White
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, U.S.A
| | - Seth M. Steinberg
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, U.S.A
| | - Leah Haworth
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, U.S.A
| | - Steven A. Rosenberg
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, U.S.A
| | - James C. Yang
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, U.S.A
| |
Collapse
|
25
|
Kumar S, Witzig TE, Rajkumar SV. Thalidomide: Current Role in the Treatment of Non-Plasma Cell Malignancies. J Clin Oncol 2004; 22:2477-88. [PMID: 15197211 DOI: 10.1200/jco.2004.10.127] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Thalidomide, initially introduced as a sedative, was withdrawn from the market in the early 1960s after it was found to be a teratogen. However, it later found use as an investigational agent in the treatment of erythema nodosum leprosum, oral ulcers, graft versus host disease, and wasting associated with the human immunodeficiency syndrome. Its antiangiogenic properties were recognized in the early 1990s during a period where the importance of angiogenesis became increasingly apparent as a critical step in the in the proliferation and spread of malignant neoplasms. This led to the evaluation of thalidomide as an antiangiogenic agent in the treatment of several cancers. Thalidomide has already become part of standard therapy for the treatment of patients with relapsed and refractory multiple myeloma. It has also been found to have varying degree of benefit in various other malignancies. Although more clinical trials are needed, Kaposi's sarcoma and myelofibrosis represent other malignancies in which thalidomide has already demonstrated promising activity. The mechanism of action of thalidomide in cancer is still unclear, but do appear to be mediated by several other mechanisms in addition to its anti-angiogenic properties. This article reviews the current status of thalidomide for the treatment of non-plasma-cell malignancies.
Collapse
Affiliation(s)
- Shaji Kumar
- Department of Internal Medicine, Mayo Clinic and Foundation, Rochester, MN 55905, USA
| | | | | |
Collapse
|
26
|
Clark PE, Hall MC, Miller A, Ridenhour KP, Stindt D, Lovato JF, Patton SE, Brinkley W, Das S, Torti FM. Phase II trial of combination interferon-alpha and thalidomide as first-line therapy in metastatic renal cell carcinoma. Urology 2004; 63:1061-5. [PMID: 15183950 DOI: 10.1016/j.urology.2004.01.035] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2003] [Accepted: 01/21/2004] [Indexed: 11/19/2022]
Abstract
OBJECTIVES To present the results of a Phase II trial of thalidomide and interferon-alpha in renal cell carcinoma. METHODS Patients with metastatic clear cell renal cell carcinoma and no prior systemic therapy were accrued. Interferon-alpha was administered at 5 million units subcutaneously three times per week. Thalidomide was started at 100 mg/day for 2 weeks and then escalated 200 mg every 2 weeks to 1000 mg or until grade 3-4 toxicity developed. Patients were assessed radiographically at baseline and after 12 weeks. Steady-state thalidomide plasma concentrations were determined. RESULTS Thirty patients were enrolled. The median age was 62 years. Seventeen patients (57%) had undergone nephrectomy before therapy. One patient died during therapy. Of the 30 patients, 29 had at least grade 2 toxicity and 17 patients had at least grade 3. At 12 weeks, no patient had a complete response, 2 had a partial response (6.7%), 8 had stable disease (26.7%), and 11 (including 1 patient with an initial partial response) had disease progression (36.7%). Nine patients were removed from the study before 12 weeks. The median follow-up was 49.6 weeks (range 2.4 to 123.7). The median time of participation in the study was 11.1 weeks (range 1.4 to 63.9). At last follow-up, 2 patients were receiving the study therapy, 1 with stable disease at 64 weeks and 1 with a partial response at 53 weeks. The median survival was 68 weeks. A linear relationship was found between the thalidomide plasma concentration and dose. No relationship was apparent between the concentration and either treatment-related toxicity or response. CONCLUSIONS Interferon-alpha and thalidomide as front-line therapy for metastatic renal cell carcinoma showed limited activity. The objective response rate was 7%. One third of patients experienced toxicity that required discontinuation of thalidomide. Randomized controlled studies are needed to determine any objective benefit of this regimen over either drug alone.
Collapse
Affiliation(s)
- Peter E Clark
- Department of Urology, Wake Forest University, Winston-Salem, North Carolina 27157, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Despite its history as a human teratogen, thalidomide is emerging as a treatment for cancer and inflammatory diseases. Although the evolution of its clinical application could not have been predicted from the tragedy associated with its misuse in the past, its history serves as a lesson in drug development that underscores the need to understand the molecular pharmacology of a compound's activity, including associated toxicities. Here, we summarise the applications for thalidomide with an emphasis on clinical trials published over the past 10 years, and consider our knowledge of the molecular pharmacology of the drug in the context of clinical trial data, attempting to provide a mechanism-guided understanding of its activity.
Collapse
Affiliation(s)
- Michael E Franks
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20030, USA
| | | | | |
Collapse
|
28
|
Abstract
PURPOSE OF REVIEW IL-2 or IFN-alpha induce remissions and prolong life in patients carefully selected for a possibly toxic treatment. However, there is a need for better-tolerated and more effective therapies, especially in patients with co-morbidities and those resistant to systemic immunotherapy. Recent achievements in the treatment of advanced renal cell carcinoma highlight potentially significant improvements. RECENT FINDINGS Cytoreductive surgery or radiation of metastases seems beneficial in well-selected patients, especially as immunotherapy is available. Immune cells within the tumour correlate with response and survival, indicating the importance of local immune modulation. Such modulation has allowed the introduction of well-tolerated treatments such as the inhalation of IL-2 to control lung metastases, which results in a significant survival benefit for high-risk patients, as suggested by a recent outcome study in 200 patients. Antibody-based tumour targeting against cG250, specifically expressed on renal cell carcinoma, seems to stabilize progressive metastatic disease and does not induce toxicity. Vaccination strategies are also well tolerated, but have not yet shown convincing results in advanced disease. Other approaches have not fulfilled expectations. Thalidomide has significant neurotoxicity and its efficacy was not confirmed in recent studies. Stem cell transplantation has significant toxicity, and cannot yet be recommended, but may have future potential. SUMMARY Cytokine-based immunotherapy can now be considered standard in the treatment of metastatic renal cell carcinoma. There is good evidence that additional local procedures such as surgery, radiation or the inhalation of IL-2 improve response and survival in metastatic disease with moderate toxicity, resulting in a significant improvement for patients suitable for these approaches.
Collapse
Affiliation(s)
- Edith Huland
- Department of Urology, University Hospital Hamburg-Eppendorf, Germany.
| | | |
Collapse
|
29
|
Hernberg M, Virkkunen P, Bono P, Ahtinen H, Mäenpää H, joensuu H. Interferon Alfa-2b Three Times Daily and Thalidomide in the Treatment of Metastatic Renal Cell Carcinoma. J Clin Oncol 2003; 21:3770-6. [PMID: 14551295 DOI: 10.1200/jco.2003.01.536] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose: The antiangiogenic effect of interferon (IFN) may improve with frequent dosing and by combination with other agents with antiangiogenic activity. To evaluate this potential, we treated patients with metastatic renal cell carcinoma (RCC) with frequently dosed IFN and thalidomide. Patients and Methods: Thirty patients were given IFN-α-2b 0.9 MU subcutaneously three times daily for 1 month and subsequently 1.2 MU tid unless serious toxicity was encountered. Thalidomide was first given 100 mg/d for 1 week and 300 mg/d thereafter. Sera were collected before and during treatment for serum vascular endothelial growth factor (S-VEGF) analyses performed using enzyme-linked immunosorbent assay. Results: The intention-to-treat response rate was 20% (95% CI, 6% to 34%) and response rate for assessable patients (n = 27) was 22% (95% CI, 6% to 38%). All responses were partial. In addition, 17 patients (63%; 95% CI, 45% to 81%) had stable disease for 3 months or longer. The median time to treatment failure was 7.7 months, and median survival time was 14.9 months. The most common cause of thalidomide discontinuation was neuropathy. S-VEGF levels decreased more in patients who responded to therapy compared with those in patients whose condition had stabilized or who had progressive disease (P = .036). Conclusion: The combination of frequently dosed IFN-α-2b and low-dose thalidomide is feasible and active in advanced RCC, but the clinical benefit may remain small compared with that of IFN alone. Results from an ongoing phase III trial comparing IFN-α with or without thalidomide need to be analyzed before this combination can be recommended for use outside clinical studies.
Collapse
Affiliation(s)
- Micaela Hernberg
- Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
| | | | | | | | | | | |
Collapse
|
30
|
Fanelli M, Sarmiento R, Gattuso D, Carillio G, Capaccetti B, Vacca A, Roccaro AM, Gasparini G. Thalidomide: a new anticancer drug? Expert Opin Investig Drugs 2003; 12:1211-25. [PMID: 12831355 DOI: 10.1517/13543784.12.7.1211] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Experimental studies have demonstrated that thalidomide (Thal), a drug developed as a sedative, has antitumoural properties. The possible antitumour mechanisms of action involve: inhibition of angiogenesis, cytokine-mediated pathways, modulation of adhesion molecules, inhibition of cyclooxygenase-2 and stimulation of immuno response. Therefore, Thal is under clinical evaluation in oncology. This paper provides an overview of the data currently available in literature regarding, in terms of activity and toxicity, the use of Thal in cancer patients. Multiple myeloma is so far the most responsive malignancy. A moderate activity has been documented in certain solid tumours: glioblastoma multiforme, renal cell carcinoma and malignant melanoma. Tolerability is generally satisfactory with peripheral neuropathy being the most relevant dose-dependent toxicity. The more frequent, but moderate side effects are: somnolence, constipation, dizziness and fatigue. More studies are needed to properly evaluate the anticancer activity of Thal alone or in combination with other anticancer treatments. Preliminary studies suggest promising results of Thal in combinations with corticosteroids and cytotoxic drugs as front-line therapy of multiple myeloma. Regarding therapy of solid tumours in the adult, combination with chemotherapy, radiation therapy and molecular-targeting compounds are under investigation.
Collapse
Affiliation(s)
- Massimo Fanelli
- Division of Clinical Oncology, San Filippo Neri Hospital, Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Takahashi M, Sugimura J, Yang X, Vogelzang N, Teh BS, Furge K, Teh BT. Gene Expression Profiling of Renal Cell Carcinoma and Its Implications in Diagnosis, Prognosis, and Therapeutics. Adv Cancer Res 2003; 89:157-81. [PMID: 14587873 DOI: 10.1016/s0065-230x(03)01005-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Renal cell carcinoma (RCC) is the 10th most common cancer in the United States. It is a histologically heterogeneous disease with various histologic types being characterized by distinct genetic alterations. This chapter reviews advances in the gene expression profiling of RCC and discusses their clinical implications. Data are promising, and many more RCC-related microarray studies are currently underway or in planning. Undoubtedly these data will have an impact on the diagnosis, prognosis, and treatment of RCCs in the future. Finally, this chapter discusses what additional studies should be performed to help uncover the molecular mechanisms of RCC and to bring this new knowledge into use in the clinical arena.
Collapse
Affiliation(s)
- Masayuki Takahashi
- Laboratory of Cancer Genetics, Van Andel Research Institute, Grand Rapids, Michigan 49503, USA
| | | | | | | | | | | | | |
Collapse
|