101
|
Fontelonga TM, Jordan B, Nunes AM, Barraza-Flores P, Bolden N, Wuebbles RD, Griner LM, Hu X, Ferrer M, Marugan J, Southall N, Burkin DJ. Sunitinib promotes myogenic regeneration and mitigates disease progression in the mdx mouse model of Duchenne muscular dystrophy. Hum Mol Genet 2020; 28:2120-2132. [PMID: 30806670 DOI: 10.1093/hmg/ddz044] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/28/2019] [Accepted: 02/18/2019] [Indexed: 02/07/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal, muscle degenerative disease causing premature death of affected children. DMD is characterized by mutations in the dystrophin gene that result in a loss of the dystrophin protein. Loss of dystrophin causes an associated reduction in proteins of the dystrophin glycoprotein complex, leading to contraction-induced sarcolemmal weakening, muscle tearing, fibrotic infiltration and rounds of degeneration and failed regeneration affecting satellite cell populations. The α7β1 integrin has been implicated in increasing myogenic capacity of satellite cells, therefore restoring muscle viability, increasing muscle force and preserving muscle function in dystrophic mouse models. In this study, we show that a Food and Drug Administration (FDA)-approved small molecule, Sunitinib, is a potent α7 integrin enhancer capable of promoting myogenic regeneration by stimulating satellite cell activation and increasing myofiber fusion. Sunitinib exerts its regenerative effects via transient inhibition of SHP-2 and subsequent activation of the STAT3 pathway. Treatment of mdx mice with Sunitinib demonstrated decreased membrane leakiness and damage owing to myofiber regeneration and enhanced support at the extracellular matrix. The decreased myofiber damage translated into a significant increase in muscle force production. This study identifies an already FDA-approved compound, Sunitinib, as a possible DMD therapeutic with the potential to treat other muscular dystrophies in which there is defective muscle repair.
Collapse
Affiliation(s)
- Tatiana M Fontelonga
- Department of Pharmacology, University of Nevada, Reno School of Medicine, , Reno, NV, USA
| | - Brennan Jordan
- Department of Pharmacology, University of Nevada, Reno School of Medicine, , Reno, NV, USA
| | - Andreia M Nunes
- Department of Pharmacology, University of Nevada, Reno School of Medicine, , Reno, NV, USA
| | - Pamela Barraza-Flores
- Department of Pharmacology, University of Nevada, Reno School of Medicine, , Reno, NV, USA
| | - Nicholas Bolden
- Department of Pharmacology, University of Nevada, Reno School of Medicine, , Reno, NV, USA
| | - Ryan D Wuebbles
- Department of Pharmacology, University of Nevada, Reno School of Medicine, , Reno, NV, USA
| | - Lesley Mathews Griner
- Division of Pre-clinical Innovation, NIH Center for Advancing Translational Sciences, Rockville, MD, USA
| | - Xin Hu
- Division of Pre-clinical Innovation, NIH Center for Advancing Translational Sciences, Rockville, MD, USA
| | - Marc Ferrer
- Division of Pre-clinical Innovation, NIH Center for Advancing Translational Sciences, Rockville, MD, USA
| | - Juan Marugan
- Division of Pre-clinical Innovation, NIH Center for Advancing Translational Sciences, Rockville, MD, USA
| | - Noel Southall
- Division of Pre-clinical Innovation, NIH Center for Advancing Translational Sciences, Rockville, MD, USA
| | - Dean J Burkin
- Department of Pharmacology, University of Nevada, Reno School of Medicine, , Reno, NV, USA
| |
Collapse
|
102
|
Brain accumulation of tivozanib is restricted by ABCB1 (P-glycoprotein) and ABCG2 (breast cancer resistance protein) in mice. Int J Pharm 2020; 581:119277. [PMID: 32234426 DOI: 10.1016/j.ijpharm.2020.119277] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/18/2020] [Accepted: 03/25/2020] [Indexed: 02/06/2023]
Abstract
Tivozanib is a potent and selective inhibitor of VEGFR1-3, recently approved by the EMA for first-line treatment of renal cell carcinoma. We used wild-type, knockout, and transgenic mouse strains to study the effects of the drug transporters ABCB1, ABCG2, and OATP1A/1B, and of the CYP3A enzymes on the oral availability and tissue distribution of tivozanib. Tivozanib was transported by human ABCB1 and mouse Abcg2 in polarized MDCK-II cells. Upon oral administration, tivozanib showed rapid absorption and the plasma concentration-time curves showed secondary peaks in all mouse strains, suggesting enterohepatic recirculation. The brain-to-plasma ratios were significantly increased in Abcb1a/1b-/- (2.2-fold) and Abcb1a/1b;Abcg2-/- (2.6-fold) mice compared to wild-type mice, indicating a modest protective role of these transporters in the blood-brain barrier. Slco1a/1b-/- mice showed a 1.2-fold lower liver-to-plasma ratio than wild-type mice, suggesting a minor role of mOatp1a/1b in tivozanib liver distribution. Oral plasma pharmacokinetics of tivozanib was not significantly altered in these mouse strains, nor in Cyp3a knockout and CYP3A4-humanized mice. The modest effect of ABC transporters on tivozanib brain accumulation, if also true in humans, might mean that this drug is not strongly limited in its therapeutic efficacy against malignant lesions situated partly or completely behind the blood-brain barrier.
Collapse
|
103
|
Bedard PL, Hyman DM, Davids MS, Siu LL. Small molecules, big impact: 20 years of targeted therapy in oncology. Lancet 2020; 395:1078-1088. [PMID: 32222192 DOI: 10.1016/s0140-6736(20)30164-1] [Citation(s) in RCA: 351] [Impact Index Per Article: 70.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 01/16/2020] [Accepted: 01/20/2020] [Indexed: 12/13/2022]
Abstract
The identification of molecular targets and the growing knowledge of their cellular functions have led to the development of small molecule inhibitors as a major therapeutic class for cancer treatment. Both multitargeted and highly selective kinase inhibitors are used for the treatment of advanced treatment-resistant cancers, and many have also achieved regulatory approval for early clinical settings as adjuvant therapies or as first-line options for recurrent or metastatic disease. Lessons learned from the development of these agents can accelerate the development of next-generation inhibitors to optimise the therapeutic index, overcome drug resistance, and establish combination therapies. The future of small molecule inhibitors is promising as there is the potential to investigate novel difficult-to-drug targets, to apply predictive non-clinical models to select promising drug candidates for human evaluation, and to use dynamic clinical trial interventions with liquid biopsies to deliver precision medicine.
Collapse
Affiliation(s)
- Philippe L Bedard
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - David M Hyman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medical College, New York, NY, USA
| | - Matthew S Davids
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Lillian L Siu
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Department of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
104
|
Deng H, Li M, Wu Q, Wang L, Hong Z, Yi F, Wei Y, Zhang W. A 2/1 Sunitinib Dosing Schedule Provides Superior Antitumor Effectiveness and Less Toxicity Than a 4/2 Schedule for Metastatic Renal Cell Carcinoma: A Systematic Review and Meta-Analysis. Front Oncol 2020; 10:313. [PMID: 32211333 PMCID: PMC7069552 DOI: 10.3389/fonc.2020.00313] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 02/21/2020] [Indexed: 01/17/2023] Open
Abstract
Background: The standard sunitinib schedule to treat metastatic renal cell carcinoma (mRCC) is 4 weeks on/2 weeks off (4/2). However, some studies revealed intolerable adverse events (AEs) in patients on this schedule. An alternative schedule, 2 weeks on/1 week off (2/1), may overcome this issue. This meta-analysis was performed to compare the effectiveness and toxicity between the 2/1 and 4/2 sunitinib dosing schedules. Methods: We acquired relevant studies by searching PubMed, ScienceDirect, the Cochrane Library, Scopus, Ovid MEDLINE, Embase, Web of Science, and Google Scholar. Our main endpoints included overall survival (OS), progression-free survival (PFS), objective response rate (ORR), disease control rate (DCR), and AEs. Results: We identified 9 medium- and high-quality studies. Both schedules were effective for mRCC, with comparable OS and similar ORR. However, the 2/1 schedule had better PFS (hazard ratio (HR) = 0.81, 95% confidence interval [CI]: 0.66–0.99, P = 0.04), higher DCR [risk rate (RR) = 1.22, 95% CI: 1.01–1.47, P = 0.04] and fewer dosage interruptions (RR = 0.60, 95% CI: 0.43–0.84, P = 0.003). Additionally, the 2/1 schedule elicited fewer specific severe AEs, including thrombocytopenia/platelet disorder, hand-foot syndrome, hypertension, and fatigue. In our subanalysis, PFS was better among East Asians using the 2/1 schedule than among other populations (HR= 0.75, 95% CI: 0.58–0.98, P = 0.03), and patients administered an initial dosage of 50 mg/d on the 2/1 schedule had superior PFS (HR = 0.76, 95% CI: 0.59–0.97, P = 0.03) than those others. Conclusions: These findings suggest that the 2/1 schedule is more suitable for mRCC than 4/2, due to superior PFS, better DCR and fewer AEs. Nevertheless, more large-scale studies with good quality are needed.
Collapse
Affiliation(s)
- Huan Deng
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Meng Li
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Qian Wu
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Li Wang
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zhengdong Hong
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Fengming Yi
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yiping Wei
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wenxiong Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
105
|
Aldemir MN, Simsek M, Kara AV, Ozcicek F, Mammadov R, Yazıcı GN, Sunar M, Coskun R, Gulaboglu M, Suleyman H. The effect of adenosine triphosphate on sunitinib-induced cardiac injury in rats. Hum Exp Toxicol 2020; 39:1046-1053. [PMID: 32131635 DOI: 10.1177/0960327120909874] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In this study, we aimed to show the effect of adenosine 5'-triphosphate (ATP) on sunitinib-induced cardiac injury in rats. The rats (n = 30) were divided equally into three groups as sunitinib group (SG), sunitinib plus ATP group (SAG), and healthy group (HG); 2 mg/kg ATP was injected intraperitoneally (ip) to the SAG group. Same volume normal saline as solvent was administered ip to the other two groups. After 1 h, 25 mg/kg sunitinib was applied orally via catheter to stomach in the SAG and SG groups. This procedure was repeated once daily for 5 weeks. At the end of this period, all animals were sacrificed and their cardiac tissue was removed. Malondialdehyde (MDA), total glutathione (tGSH), tumor necrosis factor α (TNF-α), and nuclear factor κB (NF-κB) levels in rats' cardiac tissues and troponin I (Tp-I) levels in rats' blood samples were evaluated. Histopathological analysis was also performed in cardiac tissues of the animals. MDA, TNF-α, NF-κB, and Tp-I levels were higher in the SG group compared to the SAG and HG groups (p < 0.001). tGSH levels of the SG group were lower than the SAG and HG groups (p < 0.001). The structure and morphology of cardiac muscle fibers and blood vessels were normal in the control group. In the SG group, obvious cardiac muscle tissue damage with dilated myofibers, locally atrophic myofibers, and congested blood vessels were observed. In the SAG group, marked amelioration in these findings was observed. We showed this for the first time that ATP administration exerts a protective effect against cardiac effects of sunitinib.
Collapse
Affiliation(s)
- M N Aldemir
- Department of Medical Oncology, Faculty of Medicine, Yuzuncu Yil University, Van, Turkey
| | - M Simsek
- Department of Medical Oncology, Yozgat City Hospital, Yozgat, Turkey
| | - A V Kara
- Department of Nephrology, Faculty of Medicine, Erzincan University, Erzincan, Turkey
| | - F Ozcicek
- Department of Internal Medicine, Faculty of Medicine, Erzincan University, Erzincan, Turkey
| | - R Mammadov
- Department Pharmacology, Faculty of Medicine, Erzincan University, Erzincan, Turkey
| | - G N Yazıcı
- Department of Histology and Embryology, Faculty of Medicine, Erzincan University, Erzincan, Turkey
| | - M Sunar
- Department of Anatomy, Faculty of Medicine, Erzincan University, Erzincan, Turkey
| | - R Coskun
- Department of Cardiology, Faculty of Medicine, Erzincan University, Erzincan, Turkey
| | - M Gulaboglu
- Department of Biochemistry, Faculty of Pharmacy, Ataturk University, Erzurum, Turkey
| | - H Suleyman
- Department of Pharmacology, Faculty of Medicine, Erzincan University, Erzincan, Turkey
| |
Collapse
|
106
|
Kameda T, Takayama T, Sugihara T, Takeshima S, Yamazaki M, Komatsubara M, Kamei J, Fujisaki A, Ando S, Kurokawa S, Fujimura T. The efficacy of axitinib as a first-line treatment for metastatic renal cell carcinoma. Asia Pac J Clin Oncol 2020; 16:241-246. [PMID: 32129940 DOI: 10.1111/ajco.13323] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 01/28/2020] [Indexed: 11/29/2022]
Abstract
AIM To evaluate predictive factors which associated with oncological outcomes to first-line axitinib for metastatic renal cell carcinoma (mRCC). METHODS A retrospective chart review was conducted patients who had been treated with axitinib as first-line therapy for the treatment of mRCC from September 2013 to February 2018. Axitinib was given by single daily oral administration at a dose of 10 mg, which was reduced according to adverse events (AEs). We investigated progression-free survival (PFS), overall survival (OS), objective response rate (ORR) and AEs. RESULTS Thirty-eight mRCC patients were enrolled. The median follow-up duration of axitinib treatment was 11.3 months (range = 1.0-56.9). ORR was 28.9%. Median PFS and OS was 12.8, and 17.9 months, respectively. In univariate analysis, baseline lactate dehydrogenase (LDH), neutrophil, corrected calcium (Ca), platelets (Plt) and time from diagnosis were selected as potential predictive factors. Multivariate Cox's proportional hazards model analysis showed that the number of risk factors were associated with PFS (P = 0.03) and OS (P = 0.02). CONCLUSION Baseline LDH, neutrophil, Ca, Plt and time from diagnosis are predictive factors for both PFS and OS in first-line treatment with axitinib for metastatic renal cell carcinoma.
Collapse
Affiliation(s)
- Tomohiro Kameda
- Department of Urology, Jichi Medical University, Tochigi, Japan
| | | | - Toru Sugihara
- Department of Urology, Jichi Medical University, Tochigi, Japan
| | - Saki Takeshima
- Department of Urology, Jichi Medical University, Tochigi, Japan
| | | | | | - Jun Kamei
- Department of Urology, Jichi Medical University, Tochigi, Japan
| | - Akira Fujisaki
- Department of Urology, Jichi Medical University, Tochigi, Japan
| | - Satoshi Ando
- Department of Urology, Jichi Medical University, Tochigi, Japan
| | | | | |
Collapse
|
107
|
Rizza L, Sbardella E, Gianfrilli D, Lauretta R, Tenuta M, Del Bene G, Longo F, Faggiano A, Lenzi A, Giannetta E, Pozza C. Thyroid profile during the alternative Sunitinib dosing 2/1 schedule in metastatic renal cell carcinoma. Endocrine 2020; 67:597-604. [PMID: 31679139 DOI: 10.1007/s12020-019-02088-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 09/09/2019] [Indexed: 12/18/2022]
Abstract
PURPOSE Hypothyroidism is a common side effect of Sunitinib (SUN) treatment in metastatic renal cell carcinoma (mRCC) patients. We aimed to evaluate thyroid profile during the alternative 2/1 SUN treatment schedule and to assess the predictive value of hypothyroidism in terms of survival. METHODS We performed a prospective observational study enrolling 42 consecutive mRCC patients starting first-line alternative SUN dosing 2/1 schedule. Thyroid function was assessed at baseline and during the first three SUN cycles (1 cycle = 6 weeks = 2 ON/1 OFF + 2 ON/1 OFF), and then after 6 and 12 months. Thyroid ultrasound was performed at baseline and after 3, 6, and 12 months. RESULTS Subclinical hypothyroidism developed in 24% of patients during the first cycle; in other 24% in the second cycle and in 14% in the third cycle. The highest TSH values were reached during the second cycle, ON phase (6.58 ± 5.74 μI U/l). We observed a reduction in thyroid size, in echogenicity and in parenchymal perfusion in all patients. Progression-free survival (PFS) tended to be longer in patients with TSH ≥ 5 μI U/ml during the second cycle (p = 0.069). TSH level was an independent risk factor for PFS in men (p = 0.009) but not in women (p = 0.285). CONCLUSIONS This is the first study investigating functional and morphological effects on thyroid during the alternative 2/1 SUN schedule in mRCC patients. We detected an early onset of subclinical hypothyroidism, observing the association between TSH ≥ 5 μI U/ml and: (i) longer PFS in men; (ii) progressive decrease of thyroid size in absence of significant changes in autoimmune thyroid profile.
Collapse
Affiliation(s)
- L Rizza
- Endocrinology Unit, Department of Oncology and Medical Specialities, AO San Camillo-Forlanini, Rome, Italy
| | - E Sbardella
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - D Gianfrilli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - R Lauretta
- Internal Medicine, Angiolini Hospital of Bagno di Romagna, Forlì Cesena, Italy
| | - M Tenuta
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - G Del Bene
- Department of Radiological, Oncological and Anatomopathological Sciences, Sapienza University of Rome, Rome, Italy
| | - F Longo
- Department of Radiological, Oncological and Anatomopathological Sciences, Sapienza University of Rome, Rome, Italy
| | - A Faggiano
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - A Lenzi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - E Giannetta
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - C Pozza
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
108
|
Choi SW, Lee JM, Kim DG, Noh MH. [Acute Acalculous Cholecystitis Associated with Sunitinib Treatment for Renal Cell Carcinoma]. THE KOREAN JOURNAL OF GASTROENTEROLOGY 2020; 75:103-107. [PMID: 32098465 DOI: 10.4166/kjg.2020.75.2.103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/29/2019] [Accepted: 11/20/2019] [Indexed: 11/03/2022]
Abstract
A 64-year-old man was treated with sunitinib as a first-line therapy for metastatic renal cell carcinoma. He was given oral sunitinib in cycles of 50 mg once daily for 2 weeks followed by a week off. During the 5th week of treatment right upper quadrant pain developed, but this resolved spontaneously during the 6th week (off treatment). However, on the 8th week of treatment, he was admitted to hospital because the acute right upper quadrant pain recurred with nausea, vomiting, and fever. Acute acalculous cholecystitis was then diagnosed by ultrasonography and CT. In addition, his laboratory findings indicated disseminated intravascular coagulation. Accordingly, sunitinib therapy was discontinued and broad-spectrum antibiotics initiated. He subsequently recovered after emergent percutaneous cholecystostomy. His Naranjo Adverse Drug Reaction Probability Scale score was 7, indicaing a probable association of the event with sunitinib. Suspicion of sunitinib-related acute cholecystitis is required, because, although uncommon, it can be life-threatening.
Collapse
Affiliation(s)
- Se Woong Choi
- Department of Internal Medicine, Good Gang-An Hospital, Busan, Korea
| | - Jeong Min Lee
- Department of Internal Medicine, Samyook Busan Hospital, Busan, Korea
| | - Dong Gyun Kim
- Division of Gastroenterology, Department of Internal Medicine, Dong-A University College of Medicine, Busan, Korea
| | - Myung Hwan Noh
- Division of Gastroenterology, Department of Internal Medicine, Dong-A University College of Medicine, Busan, Korea
| |
Collapse
|
109
|
Wang S, Zhu X, Han M, Hao F, Lu W, Zhou T. Mechanistic Pharmacokinetic/Pharmacodynamic Model of Sunitinib and Dopamine in MCF-7/Adr Xenografts: Linking Cellular Heterogeneity to Tumour Burden. AAPS JOURNAL 2020; 22:45. [PMID: 32043246 DOI: 10.1208/s12248-020-0428-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 01/26/2020] [Indexed: 01/08/2023]
Abstract
The self-renewal and differentiation of cancer stem-like cells (CSCs) leads to cellular heterogeneity, causing one of the greatest challenges in cancer therapy. Growing evidence suggests that CSC-targeting therapy enhances the effect of concomitant antitumour therapy. To gain an in-depth understanding of this enhanced effect, the kinetic profile of estimated CSC frequency (the fraction of CSCs in tumour) was evaluated for in vivo characterization of cellular heterogeneity using sunitinib and dopamine as a paradigm combination therapy. Female MCF-7/Adr xenografted Balb/c nude mice were treated with sunitinib (p.o., 20 mg/kg) and dopamine (i.p., 50 mg/kg), alone or in combination. Estimated CSC frequency and tumour size were measured over time. Mechanistic PK/PD modelling was performed to quantitatively describe the relationship between drug concentration, estimated CSC frequency and tumour size. Sunitinib reduced tumour size by inducing apoptosis of differentiated tumour cells (DTCs) and enriched CSCs by stimulating its proliferation. Dopamine exhibited anti-CSC effects by suppressing the capacity of CSCs and inducing its differentiation. Simulation and animal studies indicated that concurrent administration was superior to sequential administration under current experimental conditions. Alongside tumour size, the current study provides mechanistic insights into the estimation of CSC frequency as an indicator for cellular heterogeneity. This forms the conceptual basis for in vivo characterization of other combination therapies in preclinical cancer studies.
Collapse
Affiliation(s)
- Siyuan Wang
- Department of Pharmaceutics, School of Pharmaceutical sciences, Peking University, Beijing, 100191, China.,Center for Precision Medicine Multi-Omics Research, Peking University Health Science Center, Beijing, 100191, China
| | - Xiao Zhu
- Department of Pharmaceutics, School of Pharmaceutical sciences, Peking University, Beijing, 100191, China.,Otago Pharmacometrics Group, School of Pharmacy, University of Otago, Dunedin, New Zealand
| | - Mengyi Han
- Department of Pharmaceutics, School of Pharmaceutical sciences, Peking University, Beijing, 100191, China
| | - Fangran Hao
- Department of Pharmaceutics, School of Pharmaceutical sciences, Peking University, Beijing, 100191, China
| | - Wei Lu
- Department of Pharmaceutics, School of Pharmaceutical sciences, Peking University, Beijing, 100191, China.,State Key Laboratory of Natural and Biomimetic Drugs (Peking University), Beijing, 100191, China
| | - Tianyan Zhou
- Department of Pharmaceutics, School of Pharmaceutical sciences, Peking University, Beijing, 100191, China.,State Key Laboratory of Natural and Biomimetic Drugs (Peking University), Beijing, 100191, China
| |
Collapse
|
110
|
Labots M, Pham TV, Honeywell RJ, Knol JC, Beekhof R, de Goeij-de Haas R, Dekker H, Neerincx M, Piersma SR, van der Mijn JC, van der Peet DL, Meijerink MR, Peters GJ, van Grieken NC, Jiménez CR, Verheul HM. Kinase Inhibitor Treatment of Patients with Advanced Cancer Results in High Tumor Drug Concentrations and in Specific Alterations of the Tumor Phosphoproteome. Cancers (Basel) 2020; 12:cancers12020330. [PMID: 32024067 PMCID: PMC7072422 DOI: 10.3390/cancers12020330] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/20/2020] [Accepted: 01/29/2020] [Indexed: 12/22/2022] Open
Abstract
Identification of predictive biomarkers for targeted therapies requires information on drug exposure at the target site as well as its effect on the signaling context of a tumor. To obtain more insight in the clinical mechanism of action of protein kinase inhibitors (PKIs), we studied tumor drug concentrations of protein kinase inhibitors (PKIs) and their effect on the tyrosine-(pTyr)-phosphoproteome in patients with advanced cancer. Tumor biopsies were obtained from 31 patients with advanced cancer before and after 2 weeks of treatment with sorafenib (SOR), erlotinib (ERL), dasatinib (DAS), vemurafenib (VEM), sunitinib (SUN) or everolimus (EVE). Tumor concentrations were determined by LC-MS/MS. pTyr-phosphoproteomics was performed by pTyr-immunoprecipitation followed by LC-MS/MS. Median tumor concentrations were 2–10 µM for SOR, ERL, DAS, SUN, EVE and >1 mM for VEM. These were 2–178 × higher than median plasma concentrations. Unsupervised hierarchical clustering of pTyr-phosphopeptide intensities revealed patient-specific clustering of pre- and on-treatment profiles. Drug-specific alterations of peptide phosphorylation was demonstrated by marginal overlap of robustly up- and downregulated phosphopeptides. These findings demonstrate that tumor drug concentrations are higher than anticipated and result in drug specific alterations of the phosphoproteome. Further development of phosphoproteomics-based personalized medicine is warranted.
Collapse
Affiliation(s)
- Mariette Labots
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (M.L.); (T.V.P.); (R.J.H.); (J.C.K.); (R.B.); (R.d.G.-d.H.); (H.D.); (M.N.); (S.R.P.); (J.C.v.d.M.); (G.J.P.)
| | - Thang V. Pham
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (M.L.); (T.V.P.); (R.J.H.); (J.C.K.); (R.B.); (R.d.G.-d.H.); (H.D.); (M.N.); (S.R.P.); (J.C.v.d.M.); (G.J.P.)
| | - Richard J. Honeywell
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (M.L.); (T.V.P.); (R.J.H.); (J.C.K.); (R.B.); (R.d.G.-d.H.); (H.D.); (M.N.); (S.R.P.); (J.C.v.d.M.); (G.J.P.)
| | - Jaco C. Knol
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (M.L.); (T.V.P.); (R.J.H.); (J.C.K.); (R.B.); (R.d.G.-d.H.); (H.D.); (M.N.); (S.R.P.); (J.C.v.d.M.); (G.J.P.)
| | - Robin Beekhof
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (M.L.); (T.V.P.); (R.J.H.); (J.C.K.); (R.B.); (R.d.G.-d.H.); (H.D.); (M.N.); (S.R.P.); (J.C.v.d.M.); (G.J.P.)
| | - Richard de Goeij-de Haas
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (M.L.); (T.V.P.); (R.J.H.); (J.C.K.); (R.B.); (R.d.G.-d.H.); (H.D.); (M.N.); (S.R.P.); (J.C.v.d.M.); (G.J.P.)
| | - Henk Dekker
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (M.L.); (T.V.P.); (R.J.H.); (J.C.K.); (R.B.); (R.d.G.-d.H.); (H.D.); (M.N.); (S.R.P.); (J.C.v.d.M.); (G.J.P.)
| | - Maarten Neerincx
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (M.L.); (T.V.P.); (R.J.H.); (J.C.K.); (R.B.); (R.d.G.-d.H.); (H.D.); (M.N.); (S.R.P.); (J.C.v.d.M.); (G.J.P.)
| | - Sander R. Piersma
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (M.L.); (T.V.P.); (R.J.H.); (J.C.K.); (R.B.); (R.d.G.-d.H.); (H.D.); (M.N.); (S.R.P.); (J.C.v.d.M.); (G.J.P.)
| | - Johannes C. van der Mijn
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (M.L.); (T.V.P.); (R.J.H.); (J.C.K.); (R.B.); (R.d.G.-d.H.); (H.D.); (M.N.); (S.R.P.); (J.C.v.d.M.); (G.J.P.)
| | - Donald L. van der Peet
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands;
| | - Martijn R. Meijerink
- Department of Radiology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands;
| | - Godefridus J. Peters
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (M.L.); (T.V.P.); (R.J.H.); (J.C.K.); (R.B.); (R.d.G.-d.H.); (H.D.); (M.N.); (S.R.P.); (J.C.v.d.M.); (G.J.P.)
| | - Nicole C.T. van Grieken
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands;
| | - Connie R. Jiménez
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (M.L.); (T.V.P.); (R.J.H.); (J.C.K.); (R.B.); (R.d.G.-d.H.); (H.D.); (M.N.); (S.R.P.); (J.C.v.d.M.); (G.J.P.)
- Correspondence: or (C.R.J.); (H.M.W.V.)
| | - Henk M.W. Verheul
- Department of Medical Oncology, RadboudUMC, Radboud University, Geert Grooteplein Zuid 8, 6525 GA Nijmegen, The Netherlands
- Correspondence: or (C.R.J.); (H.M.W.V.)
| |
Collapse
|
111
|
Westerdijk K, Desar IME, Steeghs N, van der Graaf WTA, van Erp NP. Imatinib, sunitinib and pazopanib: From flat-fixed dosing towards a pharmacokinetically guided personalized dose. Br J Clin Pharmacol 2020; 86:258-273. [PMID: 31782166 PMCID: PMC7015742 DOI: 10.1111/bcp.14185] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/21/2019] [Accepted: 11/07/2019] [Indexed: 12/13/2022] Open
Abstract
Tyrosine kinase inhibitors (TKIs) are anti‐cancer drugs that target tyrosine kinases, enzymes that are involved in multiple cellular processes. Currently, multiple oral TKIs have been introduced in the treatment of solid tumours, all administered in a fixed dose, although large interpatient pharmacokinetic (PK) variability is described. For imatinib, sunitinib and pazopanib exposure‐treatment outcome (efficacy and toxicity) relationships have been established and therapeutic windows have been defined, therefore dose optimization based on the measured blood concentration, called therapeutic drug monitoring (TDM), can be valuable in increasing efficacy and reducing the toxicity of these drugs. In this review, an overview of the current knowledge on TDM guided individualized dosing of imatinib, sunitinib and pazopanib for the treatment of solid tumours is presented. We summarize preclinical and clinical data that have defined thresholds for efficacy and toxicity. Furthermore, PK models and factors that influence the PK of these drugs which partly explain the interpatient PK variability are summarized. Finally, pharmacological interventions that have been performed to optimize plasma concentrations are described. Based on current literature, we advise which methods should be used to optimize exposure to imatinib, sunitinib and pazopanib.
Collapse
Affiliation(s)
- Kim Westerdijk
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Ingrid M E Desar
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Neeltje Steeghs
- Department of Medical Oncology, Netherlands Cancer Institute, Antoni van Leeuwenhoek hospital, Amsterdam, the Netherlands
| | - Winette T A van der Graaf
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands.,Department of Medical Oncology, Netherlands Cancer Institute, Antoni van Leeuwenhoek hospital, Amsterdam, the Netherlands
| | - Nielka P van Erp
- Department of Clinical Pharmacy, Radboud University Medical Center, Nijmegen, the Netherlands
| | | |
Collapse
|
112
|
Huang J, Lin Y, Lai C, Yang S, Lin SF, Yang J, Huang H, Liu C, Wei W, Chuang S, Chiang C, Lee YE, Liao C, Chern CY. The inhibition profiles of 4'‐acylpyrrole–5‐fluoroindolin‐2‐ones with a C‐3' side chain for VEGFR2, PDGFR‐β, and FGFR‐1 protein kinases. J CHIN CHEM SOC-TAIP 2020. [DOI: 10.1002/jccs.201900466] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Jiann‐Jyh Huang
- Department of Applied ChemistryNational Chiayi University Chiayi City, Taiwan Republic of China
| | - Yu‐Hsiang Lin
- Development Center for BiotechnologyNational Biotechnology Research Park Taipei City, Taiwan Republic of China
| | - Chun‐Liang Lai
- Development Center for BiotechnologyNational Biotechnology Research Park Taipei City, Taiwan Republic of China
| | - Sheng‐Chuan Yang
- Development Center for BiotechnologyNational Biotechnology Research Park Taipei City, Taiwan Republic of China
| | - Shu Fu Lin
- Development Center for BiotechnologyNational Biotechnology Research Park Taipei City, Taiwan Republic of China
| | - Ju‐Ying Yang
- Development Center for BiotechnologyNational Biotechnology Research Park Taipei City, Taiwan Republic of China
| | - Hung‐Jyun Huang
- Development Center for BiotechnologyNational Biotechnology Research Park Taipei City, Taiwan Republic of China
| | - Chiawei Liu
- Development Center for BiotechnologyNational Biotechnology Research Park Taipei City, Taiwan Republic of China
| | - Win‐Yin Wei
- Development Center for BiotechnologyNational Biotechnology Research Park Taipei City, Taiwan Republic of China
| | - Shih‐Hsien Chuang
- Development Center for BiotechnologyNational Biotechnology Research Park Taipei City, Taiwan Republic of China
| | - Chao‐Cheng Chiang
- Development Center for BiotechnologyNational Biotechnology Research Park Taipei City, Taiwan Republic of China
| | - Ying‐Shuen E. Lee
- Development Center for BiotechnologyNational Biotechnology Research Park Taipei City, Taiwan Republic of China
| | - Chu‐Bin Liao
- Development Center for BiotechnologyNational Biotechnology Research Park Taipei City, Taiwan Republic of China
| | - Ching Yuh Chern
- Department of Applied ChemistryNational Chiayi University Chiayi City, Taiwan Republic of China
| |
Collapse
|
113
|
An HPLC method for simultaneous quantification of sunitinib and its active metabolite, SU12662, using hydrophilic interaction chromatography principle. Bioanalysis 2020; 12:75-85. [DOI: 10.4155/bio-2019-0188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Aim: To develop a sensitive HPLC method for the quantitation of sunitinib (SU) and its active metabolite N-desethyl-sunitinib (SU12662) in human plasma. Materials & methods: The analytes were extracted from 500 μl of plasma using liquid–liquid extraction followed by protein precipitation. Chromatographic separation of two analytes and internal standard, vandetenib, was achieved on a hydrophilic interaction liquid chromatography analytical column using a gradient program. Calibration curves were linear over the range of 10–250 ng/ml for both SU and SU12662. The method was validated according to the US FDA guidelines for bioanalytical methods. Accuracy of the method at 10 ng/ml for SU and SU12662 was 8.7 and 6.7%, respectively, and precision was 10.18% and 17.3%, respectively. Conclusion: This method allows a specific, sensitive and reliable determination of SU and SU12662 in human plasma in a single analytical run which makes it useful for therapeutic drug monitoring.
Collapse
|
114
|
Liang H, He X, Zhang Y, Chen B, Ouyang JS, Li Y, Pan B, Subba Reddy CV, Chan WTK, Qiu L. Copper-catalyzed (4+1) and (3+2) cyclizations of iodonium ylides with alkynes. Chem Commun (Camb) 2020; 56:11429-11432. [DOI: 10.1039/d0cc04373g] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The copper(ii)-catalyzed (4+1) cyclizations and copper(i)-catalyzed (3+2) cycloadditions of iodonium ylides and alkynes have been successfully developed. The corresponding highly functionalized heterocyclic products were prepared conveniently.
Collapse
|
115
|
Vercelli B, Crotti S, Agostini M. Voltammetric responses at modified electrodes and aggregation effects of two anticancer molecules: irinotecan and sunitinib. NEW J CHEM 2020. [DOI: 10.1039/d0nj03896b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Voltammetric analyses of two anticancer molecules at modified electrodes and influence of aggregate formation on their differential pulse voltammetry (DPV) responses.
Collapse
Affiliation(s)
- Barbara Vercelli
- Istituto di Chimica della Materia Condensata e di Tecnologie per l’Energia
- CNR-ICMATE
- 20125 Milano
- Italy
| | - Sara Crotti
- Istituto di Ricerca Pediatrica – Città della Speranza
- 35127 Padova
- Italy
| | - Marco Agostini
- Istituto di Ricerca Pediatrica – Città della Speranza
- 35127 Padova
- Italy
- Sezione di Clinica Chirurgica
- Dipartimento di Scienze Chirurgiche
| |
Collapse
|
116
|
Parmar A, Bjarnason GA. Individualization of Dose and Schedule Based On Toxicity for Oral VEGF Drugs in Kidney Cancer. KIDNEY CANCER 2019. [DOI: 10.3233/kca-190077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Ambika Parmar
- Sunnybrook Odette Cancer Centre, Toronto, Ontario, Canada
| | | |
Collapse
|
117
|
Medical oncology management of advanced hepatocellular carcinoma 2019: a reality check. Front Med 2019; 14:273-283. [PMID: 31863306 DOI: 10.1007/s11684-019-0728-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 10/12/2019] [Indexed: 02/06/2023]
Abstract
In terms of global cancer-related deaths, hepatocellular carcinoma (HCC) has the fourth highest mortality rate. Up until 2017, treatment of advanced HCC was largely limited to sorafenib, an oral tyrosine kinase inhibitor, with little to no success in the development of alternative treatment options. However, in the past two years, there has been an unprecedented increase in both the number and type of treatment options available for HCC. As of 2019, the US FDA has approved four oral tyrosine kinase inhibitors, two immune checkpoint inhibitors, and one anti-angiogenesis antibody for the treatment of HCC. Even with this new variety, systemic treatment of advanced HCC remains largely unsatisfactory, and the median survival rate stands at approximately one year. The expected breakthrough of using immune checkpoint inhibitors in advanced HCC did not materialize in 2019. The use of immune checkpoint inhibitors in conjunction with oral tyrosine kinase inhibitors or anti-angiogenesis medications is the current clinical research trend, the results of which are eagerly anticipated. Despite limited progress in survival, HCC research is currently experiencing a period of growth and innovation, and there is hope for significant advances in the treatment of advanced HCC as the field continues to develop.
Collapse
|
118
|
Fazio N, Cella CA, Del Re M, Laffi A, Rubino M, Zagami P, Spada F. Pharmacodynamics, clinical findings and approval status of current and emerging tyrosine-kinase inhibitors for pancreatic neuroendocrine tumors. Expert Opin Drug Metab Toxicol 2019; 15:993-1004. [PMID: 31794273 DOI: 10.1080/17425255.2019.1700951] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Introduction: Pancreatic neuroendocrine tumors (panNETs) represent a rare group of malignancies. For decades, chemotherapy, somatostatin analogs and interferon represented the only systemic therapies; however, over the latest years, new options were registered, including Everolimus, Sunitinib (SUN), and Peptide Receptor Radionuclide Therapy.Areas covered: This review discusses the role of tyrosine kinase inhibitors (TKIs) in advanced panNETs.Expert opinion: TKIs showed an antiangiogenic and antiproliferative impact on advanced panNETs. Sunitinib is the only TKI currently available in clinical practice, having been approved on the basis of relevant results of a specific panNET phase III trial. New TKIs, such as Cabozantinib, Lenvatinib, Pazopanib, Surufatinib are still on investigation in panNETs. Although some phase II studies with the new TKIs yielded better PFS and RR compared with SUN, different study designs and tumor populations may have induced selection biases. However, it was reported that panNETs resistant to SUN could respond to a new TKI, indicating a possible further therapeutic line in this context. The global investigation plan of TKIs in panNETs is not homogeneous and it is difficult to understand what kind of development this can have in the near future for clinical practice.
Collapse
Affiliation(s)
- Nicola Fazio
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO, IRCCS, Milan, Italy
| | - Chiara A Cella
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO, IRCCS, Milan, Italy
| | - Marzia Del Re
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Alice Laffi
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO, IRCCS, Milan, Italy
| | - Manila Rubino
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO, IRCCS, Milan, Italy
| | - Paola Zagami
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO, IRCCS, Milan, Italy
| | - Francesca Spada
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO, IRCCS, Milan, Italy
| |
Collapse
|
119
|
Ferreira MN, Ramseier JY, Leventhal JS. Dermatologic conditions in women receiving systemic cancer therapy. Int J Womens Dermatol 2019; 5:285-307. [PMID: 31909148 PMCID: PMC6938835 DOI: 10.1016/j.ijwd.2019.10.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 10/12/2019] [Accepted: 10/13/2019] [Indexed: 12/24/2022] Open
Abstract
As advances in cancer therapies have improved cancer-related survival, novel therapeutics have also introduced a variety of dermatologic toxicities, and an increased number of patients are living with these sequalae. Women with cancer in particular experience a spectrum of dermatologic conditions that affect their skin, hair, nail, and mucosal surfaces. Studies have shown that these toxic effects can significantly affect quality of life and alter a woman's self-image, cultural identity, femininity, sexuality, and mental health. In severe instances, dermatologic toxicities may even disrupt cancer therapy and can therefore affect overall survival and treatment response. In this article, we review the dermatologic adverse effects from traditional chemotherapy, targeted therapy, immune checkpoint inhibitors, and endocrine therapy that disproportionately affect women. The timely diagnosis and management of these dermatologic conditions is crucial in the multidisciplinary care of women with cancer.
Collapse
|
120
|
Vitiello L, Tibaudo L, Pegoraro E, Bello L, Canton M. Teaching an Old Molecule New Tricks: Drug Repositioning for Duchenne Muscular Dystrophy. Int J Mol Sci 2019; 20:E6053. [PMID: 31801292 PMCID: PMC6929176 DOI: 10.3390/ijms20236053] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/28/2019] [Accepted: 11/28/2019] [Indexed: 02/07/2023] Open
Abstract
: Duchenne muscular dystrophy (DMD) is one of the most severe forms of inherited muscular dystrophies. The disease is caused by the lack of dystrophin, a structurally essential protein; hence, a definitive cure would necessarily have to pass through some form of gene and/or cell therapy. Cell- and genetic-based therapeutics for DMD have been explored since the 1990s and recently, two of the latter have been approved for clinical use, but their efficacy is still very low. In parallel, there have been great ongoing efforts aimed at targeting the downstream pathogenic effects of dystrophin deficiency using classical pharmacological approaches, with synthetic or biological molecules. However, as it is always the case with rare diseases, R&D costs for new drugs can represent a major hurdle for researchers and patients alike. This problem can be greatly alleviated by experimenting the use of molecules that had originally been developed for different conditions, a process known as drug repurposing or drug repositioning. In this review, we will describe the state of the art of such an approach for DMD, both in the context of clinical trials and pre-clinical studies.
Collapse
Affiliation(s)
- Libero Vitiello
- Department of Biology, University of Padova, via U. Bassi 58/B, 35131 Padova, Italy;
- Interuniversity Institute of Myology (IIM), Administrative headquarters University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy;
| | - Lucia Tibaudo
- Interuniversity Institute of Myology (IIM), Administrative headquarters University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy;
- Department of Biomedical Sciences, University of Padova, via U. Bassi 58/B, 35131 Padova, Italy
| | - Elena Pegoraro
- Department of Neurosciences, University of Padova, Via Giustiniani, 5-35128 Padova, Italy;
| | - Luca Bello
- Department of Neurosciences, University of Padova, Via Giustiniani, 5-35128 Padova, Italy;
| | - Marcella Canton
- Interuniversity Institute of Myology (IIM), Administrative headquarters University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy;
- Department of Biomedical Sciences, University of Padova, via U. Bassi 58/B, 35131 Padova, Italy
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza-IRP, Corso Stati Uniti, 4, 35127 Padova, Italy
| |
Collapse
|
121
|
Ogata M, Satake H, Ogata T, Hatachi Y, Hara S, Hirota S, Yasui H. Reduction and Escalation in the Dose of Sunitinib Were Adequately Effective against Gastrointestinal Stromal Tumor of the Small Intestine. Intern Med 2019; 58:3243-3246. [PMID: 31327822 PMCID: PMC6911760 DOI: 10.2169/internalmedicine.2806-19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
We herein report the first case in which an escalated dose of sunitinib was effective, even after dose reduction. A 64-year-old man with gastrointestinal stromal tumor of the small intestine discontinued adjuvant imatinib because of interstitial pneumonia. After two years, peritoneal recurrence was detected. Sunitinib was started at 50 mg/day for 4 weeks every 6 weeks, after which the dosage was reduced to 37.5 mg/day because of grade 1 gastritis, stomatitis, and a fever. Four months later, computed tomography showed progressive disease. As the adverse events were well-controlled by medication, we escalated the dose to 50 mg/day and achieved a partial response.
Collapse
Affiliation(s)
- Misato Ogata
- Department of Medical Oncology, Kobe City Hospital Organization Kobe City Medical Center General Hospital, Japan
| | - Hironaga Satake
- Department of Medical Oncology, Kobe City Hospital Organization Kobe City Medical Center General Hospital, Japan
- Cancer Treatment Center, Kansai Medical University Hospital, Japan
| | - Takatsugu Ogata
- Department of Medical Oncology, Kobe City Hospital Organization Kobe City Medical Center General Hospital, Japan
| | - Yukimasa Hatachi
- Department of Medical Oncology, Kobe City Hospital Organization Kobe City Medical Center General Hospital, Japan
| | - Shigeo Hara
- Department of Pathology, Kobe City Hospital Organization Kobe City Medical Center General Hospital, Japan
| | - Seiichi Hirota
- Department of Surgical Pathology, Hyogo College of Medicine, Japan
| | - Hisateru Yasui
- Department of Medical Oncology, Kobe City Hospital Organization Kobe City Medical Center General Hospital, Japan
| |
Collapse
|
122
|
Jiang L, Wang L, Zhang Z, Wang Z, Wang X, Wang S, Luan X, Xia Y, Liu Y. The pharmacokinetic interaction between irinotecan and sunitinib. Cancer Chemother Pharmacol 2019; 85:443-448. [DOI: 10.1007/s00280-019-03985-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Accepted: 10/24/2019] [Indexed: 12/15/2022]
|
123
|
Fankhauser M, Bechmann N, Lauseker M, Goncalves J, Favier J, Klink B, William D, Gieldon L, Maurer J, Spöttl G, Rank P, Knösel T, Orth M, Ziegler CG, Aristizabal Prada ET, Rubinstein G, Fassnacht M, Spitzweg C, Grossman AB, Pacak K, Beuschlein F, Bornstein SR, Eisenhofer G, Auernhammer CJ, Reincke M, Nölting S. Synergistic Highly Potent Targeted Drug Combinations in Different Pheochromocytoma Models Including Human Tumor Cultures. Endocrinology 2019; 160:2600-2617. [PMID: 31322702 PMCID: PMC6795182 DOI: 10.1210/en.2019-00410] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 07/14/2019] [Indexed: 01/09/2023]
Abstract
There are no officially approved therapies for metastatic pheochromocytomas apart from ultratrace 131I-metaiodbenzylguanidine therapy, which is approved only in the United States. We have, therefore, investigated the antitumor potential of molecular-targeted approaches in murine pheochromocytoma cell lines [monocyte chemoattractant protein (MPC)/monocyte chemoattractant protein/3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT)], immortalized mouse chromaffin Sdhb-/- cells, three-dimensional pheochromocytoma tumor models (MPC/MTT spheroids), and human pheochromocytoma primary cultures. We identified the specific phosphatidylinositol-3-kinase α inhibitor BYL719 and the mammalian target of rapamycin inhibitor everolimus as the most effective combination in all models. Single treatment with clinically relevant doses of BYL719 and everolimus significantly decreased MPC/MTT and Sdhb-/- cell viability. A targeted combination of both inhibitors synergistically reduced MPC and Sdhb-/- cell viability and showed an additive effect on MTT cells. In MPC/MTT spheroids, treatment with clinically relevant doses of BYL719 alone or in combination with everolimus was highly effective, leading to a significant shrinkage or even a complete collapse of the spheroids. We confirmed the synergism of clinically relevant doses of BYL719 plus everolimus in human pheochromocytoma primary cultures of individual patient tumors with BYL719 attenuating everolimus-induced AKT activation. We have thus established a method to assess molecular-targeted therapies in human pheochromocytoma cultures and identified a highly effective combination therapy. Our data pave the way to customized combination therapy to target individual patient tumors.
Collapse
Affiliation(s)
- Maria Fankhauser
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, LMU München, Munich, Germany
| | - Nicole Bechmann
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus at Technische Universität Dresden, Dresden, Germany
| | - Michael Lauseker
- Institute for Medical Information Sciences, Biometry, and Epidemiology, Campus Grosshadern, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Judith Goncalves
- Institut National de la Santé et de la Recherche Médicale, UMR970, Paris-Cardiovascular Research Center, Equipe Labellisée par la Ligue contre le Cancer, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Judith Favier
- Institut National de la Santé et de la Recherche Médicale, UMR970, Paris-Cardiovascular Research Center, Equipe Labellisée par la Ligue contre le Cancer, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
| | - Barbara Klink
- Institute for Clinical Genetics, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
- National Center of Genetics, Laboratoire National de Santé, Dudelange, Luxembourg
- German Cancer Consortium, Dresden, Germany
| | | | - Laura Gieldon
- Institute for Clinical Genetics, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
- German Cancer Consortium, Dresden, Germany
- German Cancer Research Center, Heidelberg, Germany
- Core Unit for Molecular Tumor Diagnostics, National Center for Tumor Diseases, Heidelberg, Germany
| | - Julian Maurer
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, LMU München, Munich, Germany
| | - Gerald Spöttl
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, LMU München, Munich, Germany
| | - Petra Rank
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, LMU München, Munich, Germany
| | - Thomas Knösel
- Institute of Pathology, Ludwig-Maximilians-University, Munich, Germany
| | - Michael Orth
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Christian G Ziegler
- Department of Medicine III, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | | | - German Rubinstein
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, LMU München, Munich, Germany
| | - Martin Fassnacht
- Department of Medicine I, Division of Endocrinology and Diabetology, University Hospital, University of Würzburg, Würzburg, Germany
| | - Christine Spitzweg
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, LMU München, Munich, Germany
| | - Ashley B Grossman
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
- Royal Free Hospital ENETS Centre of Excellence, London, United Kingdom
| | - Karel Pacak
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Felix Beuschlein
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, LMU München, Munich, Germany
- Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, Universitätsspital Zürich, Zurich, Switzerland
| | - Stefan R Bornstein
- Department of Medicine III, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Graeme Eisenhofer
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus at Technische Universität Dresden, Dresden, Germany
- Department of Medicine III, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Christoph J Auernhammer
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, LMU München, Munich, Germany
| | - Martin Reincke
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, LMU München, Munich, Germany
| | - Svenja Nölting
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, LMU München, Munich, Germany
- Correspondence: Svenja Nölting, MD, Med. Klinik und Poliklinik IV, Klinikum der Universität München, Ziemssenstraße 1, 80336 Munich, Germany. E-mail:
| |
Collapse
|
124
|
Tarone L, Barutello G, Iussich S, Giacobino D, Quaglino E, Buracco P, Cavallo F, Riccardo F. Naturally occurring cancers in pet dogs as pre-clinical models for cancer immunotherapy. Cancer Immunol Immunother 2019; 68:1839-1853. [PMID: 31222484 PMCID: PMC11028358 DOI: 10.1007/s00262-019-02360-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 06/11/2019] [Indexed: 12/13/2022]
Abstract
Despite the significant progress in tumor prevention, early detection, diagnosis and treatment made over recent decades, cancer is still an enormous public health challenge all around the world, with the number of people affected increasing every year. A great deal of effort is therefore being devoted to the search for novel safe, effective and economically sustainable treatments for the growing population of neoplastic patients. One main obstacle to this process is the extremely low percentage of therapeutic approaches that, after successfully passing pre-clinical testing, actually demonstrate activity when finally tested in humans. This disappointing and expensive failure rate is partly due to the pre-clinical murine models used for in vivo testing, which cannot faithfully recapitulate the multifaceted nature and evolution of human malignancies. These features are better mirrored in natural disease models, i.e., companion animals affected by cancers. Herein, we discuss the relevance of spontaneous canine tumors for the evaluation of the safety and anti-tumor activity of novel therapeutic strategies before in-human trials, and present our experience in the development of a vaccine that targets chondroitin sulphate proteoglycan (CSPG)4 as an example of these comparative oncology studies.
Collapse
Affiliation(s)
- Lidia Tarone
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Via Nizza, 52, 10126, Turin, Italy
| | - Giuseppina Barutello
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Via Nizza, 52, 10126, Turin, Italy
| | - Selina Iussich
- Department of Veterinary Sciences, University of Turin, Largo Braccini, 2, 10095, Grugliasco, Italy
| | - Davide Giacobino
- Department of Veterinary Sciences, University of Turin, Largo Braccini, 2, 10095, Grugliasco, Italy
| | - Elena Quaglino
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Via Nizza, 52, 10126, Turin, Italy
| | - Paolo Buracco
- Department of Veterinary Sciences, University of Turin, Largo Braccini, 2, 10095, Grugliasco, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Via Nizza, 52, 10126, Turin, Italy.
| | - Federica Riccardo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Via Nizza, 52, 10126, Turin, Italy
| |
Collapse
|
125
|
Marangon E, Buzzo M, Posocco B, Gagno S, Zanchetta M, Iacuzzi V, Poetto AS, Guardascione M, Giodini L, Toffoli G. A new high-performance liquid chromatography-tandem mass spectrometry method for the determination of sunitinib and N-desethyl sunitinib in human plasma: Light-induced isomerism overtaking towards therapeutic drug monitoring in clinical routine. J Pharm Biomed Anal 2019; 179:112949. [PMID: 31784210 DOI: 10.1016/j.jpba.2019.112949] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 10/14/2019] [Accepted: 10/21/2019] [Indexed: 02/02/2023]
Abstract
Sunitinib is approved for advanced renal cell cancer, imatinib-resistant or -intolerant gastrointestinal stromal tumors and pancreatic neuroendocrine cancers. It is prescribed at a fixed dose but its plasma exposure shows large inter-individual variations. Taking into account the narrow therapeutic window and the positive exposure-efficacy relationship, there is a robust rationale for its therapeutic drug monitoring. In fact, a target plasma concentration of sunitinib plus its active metabolite, N-desethyl sunitinib, ≥50 ng/mL was suggested. In order to quantify sunitinib and N-desethyl sunitinib in patients' plasma, we developed and validated a new LC-MS/MS method applicable to clinical routine. In solution, sunitinib and N-desethyl sunitinib undergo to photo-isomerization and many published methods overcome this problem by conducting the entire procedures of samples collection and handling under strictly light-protection. Our method is based on a simple and fast procedure that quantitatively reconverts the E-isomer of both analytes, obtained during sample draw and processing without light-protection, into their Z-forms. Moreover, our method uses a small plasma volume (30 μL) and the analytes are extracted by a rapid protein precipitation. It was validated according to EMA-FDA guidelines. The calibration curves resulted linear (R2 always >0.993) over the concentration ranges (0.1-500 ng/mL for sunitinib, 0.1-250 ng/mL for N-desethyl sunitinib) with a good precision (within 7.7 % for sunitinib and 10.8% for N- desethyl sunitinib) and accuracy (range 95.8-102.9% for sunitinib and 92.3-106.2% for N-desethyl sunitinib). This method was applied to a pharmacokinetic study in one patient treated with sunitinib. Moreover, as incurred samples reanalysis is an established part of the bioanalytical process to support clinical studies, its assessment was performed early in order to assure that any reproducibility issues was detected as soon as possible. The percentage difference between the two runs resulted within ±20% in all the re-analysed samples for both sunitinib and N- desethyl sunitinib.
Collapse
Affiliation(s)
- Elena Marangon
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Pordenone, Italy.
| | - Mauro Buzzo
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Pordenone, Italy
| | - Bianca Posocco
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Pordenone, Italy
| | - Sara Gagno
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Pordenone, Italy
| | - Martina Zanchetta
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Pordenone, Italy
| | - Valentina Iacuzzi
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Pordenone, Italy
| | - Ariana Soledad Poetto
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Pordenone, Italy
| | - Michela Guardascione
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Pordenone, Italy
| | - Luciana Giodini
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Pordenone, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Pordenone, Italy
| |
Collapse
|
126
|
Pozas J, San Román M, Alonso-Gordoa T, Pozas M, Caracuel L, Carrato A, Molina-Cerrillo J. Targeting Angiogenesis in Pancreatic Neuroendocrine Tumors: Resistance Mechanisms. Int J Mol Sci 2019; 20:E4949. [PMID: 31597249 PMCID: PMC6801829 DOI: 10.3390/ijms20194949] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/04/2019] [Accepted: 10/05/2019] [Indexed: 02/07/2023] Open
Abstract
Despite being infrequent tumors, the incidence and prevalence of pancreatic neuroendocrine tumors (P-NETs) has been rising over the past few decades. In recent years, rigorous phase III clinical trials have been conducted, allowing the approval of several drugs that have become the standard of care in these patients. Although various treatments are used in clinical practice, including somatostatin analogues (SSAs), biological therapies like sunitinib or everolimus, peptide receptor radionuclide therapy (PRRT) or even chemotherapy, a consensus regarding the optimal sequence of treatment has not yet been reached. Notwithstanding, sunitinib is largely used in these patients after the promising results shown in SUN111 phase III clinical trial. However, both prompt progression as well as tumor recurrence after initial response have been reported, suggesting the existence of primary and acquired resistances to this antiangiogenic drug. In this review, we aim to summarize the most relevant mechanisms of angiogenesis resistance that are key contributors of tumor progression and dissemination. Furthermore, several targeted molecules acting selectively against these pathways have shown promising results in preclinical models, and preliminary results from ongoing clinical trials are awaited.
Collapse
Affiliation(s)
- Javier Pozas
- Medical Oncology Department, University Hospital Ramon y Cajal, 28034 Madrid, Spain.
| | - María San Román
- Medical Oncology Department, University Hospital Ramon y Cajal, 28034 Madrid, Spain.
| | - Teresa Alonso-Gordoa
- Medical Oncology Department, University Hospital Ramon y Cajal, 28034 Madrid, Spain.
- The Ramón y Cajal Health Research Institute (IRYCIS), CIBERONC, 28034 Madrid, Spain.
- Alcalá University, 28805 Madrid, Spain.
| | - Miguel Pozas
- Medical Oncology Department, University Hospital Ramon y Cajal, 28034 Madrid, Spain.
| | - Laura Caracuel
- Medical Oncology Department, University Hospital Ramon y Cajal, 28034 Madrid, Spain.
| | - Alfredo Carrato
- Medical Oncology Department, University Hospital Ramon y Cajal, 28034 Madrid, Spain.
- The Ramón y Cajal Health Research Institute (IRYCIS), CIBERONC, 28034 Madrid, Spain.
- Alcalá University, 28805 Madrid, Spain.
| | - Javier Molina-Cerrillo
- Medical Oncology Department, University Hospital Ramon y Cajal, 28034 Madrid, Spain.
- The Ramón y Cajal Health Research Institute (IRYCIS), CIBERONC, 28034 Madrid, Spain.
- Alcalá University, 28805 Madrid, Spain.
| |
Collapse
|
127
|
Corbaux P, El-Madani M, Tod M, Péron J, Maillet D, Lopez J, Freyer G, You B. Clinical efficacy of the optimal biological dose in early-phase trials of anti-cancer targeted therapies. Eur J Cancer 2019; 120:40-46. [DOI: 10.1016/j.ejca.2019.08.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 07/26/2019] [Accepted: 08/02/2019] [Indexed: 10/26/2022]
|
128
|
London AJ, Kimmelman J. Clinical Trial Portfolios: A Critical Oversight in Human Research Ethics, Drug Regulation, and Policy. Hastings Cent Rep 2019; 49:31-41. [DOI: 10.1002/hast.1034] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
129
|
Bruchbacher A, Nachbargauer S, Fajkovic H, Schmidinger M. Sunitinib Dose Escalation in Metastatic Renal Cell Carcinoma. KIDNEY CANCER 2019. [DOI: 10.3233/kca-190055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Andreas Bruchbacher
- Department of Medicine I, Clinical Division of Oncology and Comprehensive Cancer Center, Medical University of Vienna, Austria
| | - Sebastian Nachbargauer
- Department of Medicine I, Clinical Division of Oncology and Comprehensive Cancer Center, Medical University of Vienna, Austria
| | - Harun Fajkovic
- Department of Urology, Medical University of Vienna, Austria
| | - Manuela Schmidinger
- Department of Medicine I, Clinical Division of Oncology and Comprehensive Cancer Center, Medical University of Vienna, Austria
| |
Collapse
|
130
|
Zhao Q, Zhang T, Xiao X, Huang J, Wang Y, Gonzalez FJ, Li F. Impaired clearance of sunitinib leads to metabolic disorders and hepatotoxicity. Br J Pharmacol 2019; 176:2162-2178. [PMID: 30875096 PMCID: PMC6555861 DOI: 10.1111/bph.14664] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 01/23/2019] [Accepted: 03/05/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE Sunitinib is a small-molecule TK inhibitor associated with hepatotoxicity. The mechanisms of its toxicity are still unclear. EXPERIMENTAL APPROACH In the present study, mice were treated with 60, 150, and 450 mg·kg-1 sunitinib to evaluate sunitinib hepatotoxicity. Sunitinib metabolites and endogenous metabolites in liver, serum, faeces, and urine were analysed using ultra-performance LC electrospray ionization quadrupole time-of-flight MS-based metabolomics. KEY RESULTS Four reactive metabolites and impaired clearance of sunitinib in liver played a dominant role in sunitinib-induced hepatotoxicity. Using a non-targeted metabolomics approach, various metabolic pathways, including mitochondrial fatty acid β-oxidation (β-FAO), bile acids, lipids, amino acids, nucleotides, and tricarboxylic acid cycle intermediates, were disrupted after sunitinib treatment. CONCLUSIONS AND IMPLICATIONS These studies identified significant alterations in mitochondrial β-FAO and bile acid homeostasis. Activation of PPARα and inhibition of xenobiotic metabolism may be of value in attenuating sunitinib hepatotoxicity.
Collapse
Affiliation(s)
- Qi Zhao
- State Key Laboratory of Phytochemistry and Plant Resources in West China Kunming Institute of BotanyChinese Academy of SciencesKunmingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Ting Zhang
- State Key Laboratory of Phytochemistry and Plant Resources in West China Kunming Institute of BotanyChinese Academy of SciencesKunmingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Xue‐Rong Xiao
- State Key Laboratory of Phytochemistry and Plant Resources in West China Kunming Institute of BotanyChinese Academy of SciencesKunmingChina
| | - Jian‐Feng Huang
- State Key Laboratory of Phytochemistry and Plant Resources in West China Kunming Institute of BotanyChinese Academy of SciencesKunmingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yan Wang
- Department of PathologyThe Second Affiliated Hospital of Kunming Medical UniversityKunmingChina
| | - Frank J. Gonzalez
- Laboratory of Metabolism, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMD
| | - Fei Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China Kunming Institute of BotanyChinese Academy of SciencesKunmingChina
- Jiangxi University of Traditional Chinese MedicineNanchangChina
| |
Collapse
|
131
|
Chen C, Fang H, Jiao Y, Zhou Y, Guo Q, Lv Z. Clinical Efficacy and Complication Rate of Sunitinib 2/1 Versus 4/2 Schedule for the Treatment of Metastatic Renal Cell Cancer: A Systematic Review and Meta-Analysis. Clin Genitourin Cancer 2019; 17:319-331. [PMID: 31371223 DOI: 10.1016/j.clgc.2019.06.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/26/2019] [Accepted: 06/03/2019] [Indexed: 12/24/2022]
Abstract
The treatment of renal cell carcinoma has achieved certain curative effects with the innovation of clinical drugs, such as sunitinib. However, the clinical efficacy and complication rate of the sunitinib 2/1 and 4/2 schedule in metastatic renal cell cancer remain unclear. In this study we aimed to resolve this issue by using meta-analysis to provide more theoretical guidance for clinical use. Several outcome measurements were included in this study to compare the 2 schedules such as complete response, partial response, stable disease, progressive disease, progression-free survival, overall survival, and complications. In the contrast analysis, the sunitinib 2/1 and 4/2 schedule resulted in significant improvements in prognosis. However, the sunitinib 2/1 schedule was superior to the 4/2 schedule in terms of controlling stable disease and causing fewer complications.
Collapse
Affiliation(s)
- Chenglong Chen
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Taiyuan, P.R. China
| | - Huan Fang
- Urology Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, P.R. China
| | - Yurui Jiao
- Department of Endocrinology, The Second Hospital of Shanxi Medical University, Taiyuan, P.R. China
| | - Yi Zhou
- First Clinical Medical School of Southern Medical University, Guangzhou, P.R. China
| | - Qiang Guo
- Urology Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, P.R. China
| | - Zhi Lv
- Department of Endocrinology, The Second Hospital of Shanxi Medical University, Taiyuan, P.R. China.
| |
Collapse
|
132
|
Yamaguchi H, Takasaki S, Kikuchi M, Kawasaki Y, Arai Y, Mano N. [Toward Personalized Cancer Therapy with Oral Molecular-targeted Agents]. YAKUGAKU ZASSHI 2019; 139:911-915. [PMID: 31155535 DOI: 10.1248/yakushi.18-00213-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Oral molecular-targeted agents are used clinically for the treatment of various types of cancer. However, even when treatment is started at the dosage indicated in the medical package insert, we have experienced many cases in which treatment had to be stopped early owing to the occurrence of serious side effects or an insufficient therapeutic effect. In recent years, a wide range of studies has been conducted on the therapeutic drug monitoring (TDM) of oral molecular-targeted therapeutic agents to prevent serious side effects and maximize the therapeutic effect. In Japan, the TDM of imatinib has been covered by insurance since 2012, and the TDM of sunitinib has been covered since 2018. In contrast, tyrosine kinase inhibitors may have severe side effects, but their TDM is not covered by medical insurance. We aimed to identify a safe, highly effective chemotherapy regimen based on scientific evidence gathered from Japanese patients. We examined the relationship between the plasma concentration of drugs and clinical findings, such as side effects and treatment effects, at our hospital. In this symposium review, we introduce our results based on the treatment of patients with renal cell carcinoma.
Collapse
Affiliation(s)
| | - Shinya Takasaki
- Department of Pharmaceutical Sciences, Tohoku University Hospital
| | - Masafumi Kikuchi
- Department of Pharmaceutical Sciences, Tohoku University Hospital
| | | | - Yoichi Arai
- Department of Urology, Tohoku University Hospital
| | - Nariyasu Mano
- Department of Pharmaceutical Sciences, Tohoku University Hospital
| |
Collapse
|
133
|
Bresciani G, Ditsiou A, Cilibrasi C, Vella V, Rea F, Schiavon M, Cavallesco NG, Giamas G, Zatelli MC, Gagliano T. EGF and IGF1 affect sunitinib activity in BP-NEN: new putative targets beyond VEGFR? Endocr Connect 2019; 8:680-690. [PMID: 31035254 PMCID: PMC6528406 DOI: 10.1530/ec-19-0192] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 04/29/2019] [Indexed: 02/06/2023]
Abstract
Broncho-pulmonary neuroendocrine neoplasms (BP-NENs) are neoplasms orphan of an efficient therapy. Available medical treatments derived from clinical trials are not specific for the management of this malignancy. Sunitinib is a multi-receptor tyrosine-kinases (RTKs) inhibitor that has already shown its efficacy in NENs, but there are no available data about its action in BP-NENs. Therefore, our aim was to understand the effects of RTKs inhibition promoted by sunitinib in order to evaluate new putative targets useful in malignancy treatment. Since our results underlined a role for EGFR and IGF1R in modulating sunitinib antiproliferative action, we investigated the effects of erlotinib, an EGFR inhibitor, and linsitinib, an IGF1R inhibitor, in order to understand their function in regulating cells behaviour. Cell viability and caspase activation were evaluated on two immortalised human BP-NEN cell lines and primary cultures. Our results showed that after treatment with sunitinib and/or IGF1, EGF and VEGF, the antiproliferative effect of sunitinib was counteracted by EGF and IGF1 but not by VEGF. Therefore, we evaluated with AlphaScreen technology the phosphorylated EGFR and IGF1R levels in primary cultures treated with sunitinib and/or EGF and IGF1. Results showed a decrease of p-IGF1R after treatment with sunitinib and an increase after co-treatment with IGF1. Then, we assessed cell viability and caspase activation on BP-NEN cell lines after treatment with linsitinib and/or erlotinib. Results demonstrate that these two agents have a stronger antiproliferative effect compared to sunitinib. In conclusion, our results suggest that IGF1R and EGF1R could represent putative molecular targets in BP-NENs treatment.
Collapse
Affiliation(s)
- Giulia Bresciani
- Section of Endocrinology and Internal Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Angeliki Ditsiou
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, UK
| | - Chiara Cilibrasi
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, UK
| | - Viviana Vella
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, UK
| | - Federico Rea
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Padua, Italy
| | - Marco Schiavon
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Padua, Italy
| | | | - Georgios Giamas
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, UK
| | - Maria Chiara Zatelli
- Section of Endocrinology and Internal Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Teresa Gagliano
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, UK
- Correspondence should be addressed to T Gagliano:
| |
Collapse
|
134
|
Llovet JM, Montal R, Villanueva A. Randomized trials and endpoints in advanced HCC: Role of PFS as a surrogate of survival. J Hepatol 2019; 70:1262-1277. [PMID: 30943423 DOI: 10.1016/j.jhep.2019.01.028] [Citation(s) in RCA: 156] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 12/21/2018] [Accepted: 01/29/2019] [Indexed: 02/08/2023]
Abstract
Hepatocellular carcinoma (HCC) is a major cause of cancer-related mortality worldwide. Around half of patients with HCC will receive systemic therapies during their life span. The pivotal positive sorafenib trial and regulatory approval in 2007 was followed by a decade of negative studies with drugs leading to marginal antitumoral efficacy, toxicity, or trials with a lack of enrichment strategies. This trend has changed over the last 2 years with several compounds, such as lenvatinib (in first-line) and regorafenib, cabozantinib, ramucirumab and nivolumab (in second-line), showing clinical benefit. These successes came at a cost of increasing the complexity of decision-making, and ultimately, impacting the design of future clinical trials. Nowadays, life expectancy with single active agents has surpassed the threshold of 1 year and sequential strategies have provided encouraging outcomes. Overall survival (OS) remains the main endpoint in phase III investigations, but as in other solid tumours, there is a clear need to define surrogate endpoints that both reliably recapitulate survival benefits and can be assessed before additional efficacious drugs are administered. A thorough analysis of 21 phase III trials published in advanced HCC demonstrated a moderate correlation between progression-free survival (PFS) or time to progression (TTP) and OS (R = 0.84 and R = 0.83, respectively). Nonetheless, the significant differences in PFS identified in 7 phase III studies only correlated with differences in OS in 3 cases. In these cases, the hazard ratio (HR) for PFS was ≤0.6. Thus, this threshold is herein proposed as a potential surrogate endpoint of OS in advanced HCC. Conversely, PFS with an HR between 0.6-0.7, despite significance, was not associated with better survival, and thus these magnitudes are considered uncertain surrogates. In the current review, we discuss the reasons for positive or negative phase III trials in advanced HCC, and the strengths and limitations of surrogate endpoints (PFS, TTP and objective response rate [ORR]) to predict survival.
Collapse
Affiliation(s)
- Josep M Llovet
- Translational Research in Hepatic Oncology, Liver Unit, IDIBAPS, Hospital Clinic Barcelona, University of Barcelona, Barcelona, Catalonia, Spain; Mount Sinai Liver Cancer Program, Division of Liver Diseases, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain.
| | - Robert Montal
- Translational Research in Hepatic Oncology, Liver Unit, IDIBAPS, Hospital Clinic Barcelona, University of Barcelona, Barcelona, Catalonia, Spain
| | - Augusto Villanueva
- Mount Sinai Liver Cancer Program, Division of Liver Diseases, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
135
|
Sun Y, Li J, Yang X, Zhang G, Fan X. The Alternative 2/1 Schedule of Sunitinib is Superior to the Traditional 4/2 Schedule in Patients With Metastatic Renal Cell Carcinoma: A Meta-analysis. Clin Genitourin Cancer 2019; 17:e847-e859. [PMID: 31279483 DOI: 10.1016/j.clgc.2019.05.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 03/21/2019] [Accepted: 05/09/2019] [Indexed: 01/10/2023]
Abstract
Alternate sunitinib schedules attracted the interest of oncologists recently owing to their superior safety profile. This meta-analysis compared the tolerability and efficacy of a new alternative dosing schedule (2 weeks on/1 week off) of sunitinib with the traditional 4/2 schedule in patients with metastatic renal cell carcinoma (mRCC). Studies were retrieved from Medline, Cochrane Central, Scopus, Embase, and Web of Science databases. Data were extracted and pooled as hazard ratio (HR: survival data) or odds ratio (OR: dichotomous data) using Comprehensive Meta-analysis software. Based on data of 1173 patients, the progression-free survival (HR, 0.52; 95% confidence interval [CI], 0.39-0.95; P < .0001), overall survival (HR, 0.6; 95% CI, 0.43-0.85; P < .0001), and stable disease rates (OR, 0.38; 95% CI, 0.19-0.76; P = .006) were significantly improved on the alternative 2/1 schedule, compared with the traditional 4/2 schedule. However, the complete response (OR, 1.32; 95% CI, 0.34-5.22; P = .69) and partial response (OR, 1.34; 95% CI, 0.44-4.14; P = .61) rates were comparable between the 2 regimens. The tolerability of the alternative 2/1 schedule was superior to the traditional one as investigated adverse events like fatigue (OR, 2.91; 95% CI, 1.89-4.46; P < .0001), hypertension (OR, 2.08; 95% CI, 1.56-2.75; P < .0001), and diarrhea (OR, 2.18; 95% CI, 1.19-3.98; P = .012) were significantly less common. In conclusion, the alternative 2/1 sunitinib schedule provides improved tolerability and survival in patients with mRCC. Large randomized trials with long follow-up periods are required to validate and confirm these findings.
Collapse
Affiliation(s)
- Yi Sun
- Department of Laboratory Medicine, The First People's Hospital of Yunnan Province, Yunnan, Kunming, Yunnan Province, China
| | - Jiejing Li
- Department of Laboratory Medicine, The First People's Hospital of Yunnan Province, Yunnan, Kunming, Yunnan Province, China
| | - Xiangcai Yang
- Department of Laboratory Medicine, The First People's Hospital of Yunnan Province, Yunnan, Kunming, Yunnan Province, China
| | - Guiqian Zhang
- Department of Laboratory Medicine, The First People's Hospital of Yunnan Province, Yunnan, Kunming, Yunnan Province, China
| | - Xin Fan
- Department of Laboratory Medicine, The First People's Hospital of Yunnan Province, Yunnan, Kunming, Yunnan Province, China.
| |
Collapse
|
136
|
Pokuri VK, Tomaszewski GM, Ait-Oudhia S, Groman A, Khushalani NI, Lugade AA, Thanavala Y, Ashton EA, Grande C, Fetterly GJ, Iyer R. Efficacy, Safety, and Potential Biomarkers of Sunitinib and Transarterial Chemoembolization (TACE) Combination in Advanced Hepatocellular Carcinoma (HCC): Phase II Trial. Am J Clin Oncol 2019; 41:332-338. [PMID: 27014931 DOI: 10.1097/coc.0000000000000286] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVES To evaluate the safety/efficacy and explore biomarkers for a rationally designed combination of sunitinib and transarterial chemoembolization (TACE) in a prospective phase 2 study of advanced hepatocellular carcinoma (HCC). METHODS Inoperable HCC patients with Child-Pugh A disease received 37.5 mg sunitinib from days 1 to 7 followed by TACE on day 8. Sunitinib was resumed from days 15 to 36 followed by 2 weeks off. Patients received subsequent sunitinib cycles of 4 weeks on and 2 weeks off. Dynamic contrast-enhanced magnetic resonance imaging and circulating soluble biomarkers were assessed at baseline, day 8, day 10, and day 36. RESULTS Sixteen patients with liver only (n=10) and extrahepatic disease (n=6) were enrolled. After a median follow-up of 12.8 months, 2 partial responses, 11 stable disease, and 3 clinical deteriorations were seen for a clinical benefit rate of 81%. Median progression-free survival (PFS) was 8 months (95% CI, 4.3-9.3) and overall survival was 14.9 months (95% CI, 6.3-27.1). Eleven of 16 patients (69%) had grade 3/4 toxicities attributable to sunitinib, the most frequent being thrombocytopenia, amylase/lipase elevations, lymphopenia, and fatigue. Mean K (volume transfer constant) and viable tumor percent in consented patients decreased by 27% and 14.8%, respectively, with combination therapy. Soluble vascular endothelial growth factor receptor-2 (sVEGFR2) levels, cytokines (interleukin-8, interleukin-21), and monocytes decreased with combination therapy. Estimated sunitinib IC50 values of 15 and 10 ng/mL modulated K and AUC90. sVEGFR2 levels decreased with K and AUC90. CONCLUSIONS Encouraging progression-free survival and overall survival were seen with acceptable toxicity in our study of sunitinib and TACE combination in advanced HCC. Potential imaging and serum biomarkers showed increased benefit with combination therapy.
Collapse
Affiliation(s)
| | | | - Sihem Ait-Oudhia
- Pharmacometrics and Systems Pharmacology at Lake Nona, University of Florida, Orlando, FL
| | | | | | - Amit A Lugade
- Center for Immunotherapy, Roswell Park Cancer Institute (RPCI), Buffalo
| | | | | | | | | | | |
Collapse
|
137
|
Li C, Zou R, Zhang H, Wang Y, Qiu B, Qiu S, Wang W, Xu Y. Upregulation of phosphoinositide 3-kinase prevents sunitinib-induced cardiotoxicity in vitro and in vivo. Arch Toxicol 2019; 93:1697-1712. [DOI: 10.1007/s00204-019-02448-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 04/09/2019] [Indexed: 12/20/2022]
|
138
|
Alonso-Gordoa T, García-Bermejo ML, Grande E, Garrido P, Carrato A, Molina-Cerrillo J. Targeting Tyrosine kinases in Renal Cell Carcinoma: "New Bullets against Old Guys". Int J Mol Sci 2019; 20:E1901. [PMID: 30999623 PMCID: PMC6515337 DOI: 10.3390/ijms20081901] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 04/14/2019] [Accepted: 04/15/2019] [Indexed: 12/24/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the seventh most frequently diagnosed tumor in adults in Europe and represents approximately 2.5% of cancer deaths. The molecular biology underlying renal cell carcinoma (RCC) development and progression has been a key milestone in the management of this type of tumor. The discovery of Von Hippel Lindau (VHL) gene alterations that arouse in 50% of ccRCC patients, leads the identification of an intracellular accumulation of HIF and, consequently an increase of VEGFR expression. This change in cell biology represents a new paradigm in the treatment of metastatic renal cancer by targeting angiogenesis. Currently, there are multiple therapeutic drugs available for advanced disease, including therapies against VEGFR with successful results in patients´ survival. Other tyrosine kinases' pathways, including PDGFR, Axl or MET have emerged as key signaling pathways involved in RCC biology. Indeed, promising new drugs targeting those tyrosine kinases have exhibited outstanding efficacy. In this review we aim to present an overview of the central role of these tyrosine kinases' activities in relevant biological processes for kidney cancer and their usefulness in RCC targeted therapy development. In the immunotherapy era, angiogenesis is still an "old guy" that the medical community is trying to fight using "new bullets".
Collapse
Affiliation(s)
- Teresa Alonso-Gordoa
- Medical Oncology Department, The Ramón y Cajal Health Research Institute (IRYCIS), CIBERONC, Alcalá University, University Hospital Ramon y Cajal, 28034 Madrid, Spain.
| | - María Laura García-Bermejo
- Biomarkers and Therapeutic Targets Group and Core Facility, Ramón y Cajal Research Institute, (IRYCIS), 28034 Madrid, Spain.
| | - Enrique Grande
- Medical Oncology Department, MD Anderson Cancer Center, 28034 Madrid, Spain.
| | - Pilar Garrido
- Medical Oncology Department, The Ramón y Cajal Health Research Institute (IRYCIS), CIBERONC, Alcalá University, University Hospital Ramon y Cajal, 28034 Madrid, Spain.
| | - Alfredo Carrato
- Medical Oncology Department, Ramón y Cajal Health Research Institute (IRYCIS). CIBERONC, Alcalá University, University Hospital Ramon y Cajal, 28034 Madrid, Spain.
| | - Javier Molina-Cerrillo
- Medical Oncology Department, The Ramón y Cajal Health Research Institute (IRYCIS), CIBERONC, Alcalá University, University Hospital Ramon y Cajal, 28034 Madrid, Spain.
| |
Collapse
|
139
|
Valle JW, Borbath I, Rosbrook B, Fernandez K, Raymond E. Sunitinib in patients with pancreatic neuroendocrine tumors: update of safety data. Future Oncol 2019; 15:1219-1230. [DOI: 10.2217/fon-2018-0882] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Aim: To describe the long-term safety of sunitinib in patients with progressive, well-differentiated, advanced/metastatic pancreatic neuroendocrine tumors. Patients & methods: Sunitinib- and placebo-treated patients from the Phase III study continued to receive sunitinib (37.5 mg on a continuous daily-dosing regimen) in two open-label extension studies. Results: Median (range) treatment exposure: 30.2 (0.7–269.4) and 87.1 (3.9–319.4) weeks for medium-term (n = 41) and long-term-treated (n = 61) populations, respectively. All patients experienced ≥1 adverse event (AE); 47 (45.6%) reported serious AEs. Common all-causality AEs: diarrhea (63.1%); neutropenia (43.7%); abdominal pain (40.8%). Fifteen (14.6%) patients discontinued treatment due to treatment-related AEs. Conclusion: The safety of extended sunitinib treatment was consistent with the known safety profile of sunitinib in pancreatic neuroendocrine tumors.
Collapse
Affiliation(s)
- Juan W Valle
- Division of Cancer Sciences/Department of Medical Oncology, The Christie NHS Foundation Trust, University of Manchester, Manchester M20 4BX, UK
| | - Ivan Borbath
- Department of Hepato-Gastroenterology, Cliniques Universitaires Saint-Luc, Av Hippocrate, 10 - 1200 Brussels, Belgium
| | - Brad Rosbrook
- Pfizer Inc., 10646 Science Center Dr, San Diego, CA 92121, USA
| | | | - Eric Raymond
- Paris Saint-Joseph Hospital Group, 185 Rue Raymond Losserand, Paris 75014, France
| |
Collapse
|
140
|
Calvo E, Porta C, Grünwald V, Escudier B. The Current and Evolving Landscape of First-Line Treatments for Advanced Renal Cell Carcinoma. Oncologist 2019; 24:338-348. [PMID: 30158285 PMCID: PMC6519762 DOI: 10.1634/theoncologist.2018-0267] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 07/05/2018] [Indexed: 11/17/2022] Open
Abstract
Agents targeting the vascular endothelial growth factor (VEGF) and its receptors (VEGFRs), as well as the mammalian target of rapamycin (mTOR) and immune checkpoint receptor programmed death 1 (PD-1) signaling pathway have improved clinical outcomes for patients with advanced renal cell carcinoma (RCC). The VEGFR tyrosine kinase inhibitors (TKIs) pazopanib and sunitinib are FDA-approved first-line treatment options for advanced RCC; however, other treatment options in this setting are available, including the recently approved combination of nivolumab (anti-PD-1) and ipilimumab (anti-cytotoxic T-lymphocyte-associated protein-4 [CTLA-4]) for patients with intermediate or poor risk. Unfortunately, treatment guideline recommendations provide little guidance to aid first-line treatment choice. In addition, several ongoing randomized phase III trials of investigational first-line regimens may complicate the RCC treatment paradigm if these agents gain approval. This article reviews clinical trial and real-world evidence for currently approved and investigational first-line treatment regimens for advanced RCC and provides clinical evidence to aid first-line treatment selection. IMPLICATIONS FOR PRACTICE: Vascular endothelial growth factor receptor tyrosine kinase inhibitors are approved by the U.S. Food and Drug Administration as first-line treatment options for advanced renal cell carcinoma; however, the treatment paradigm is rapidly evolving. The combination of nivolumab plus ipilimumab was recently approved for intermediate- and poor-risk patients, and other combination strategies and novel first-line agents will likely be introduced soon.
Collapse
Affiliation(s)
- Emiliano Calvo
- Centro Integral Oncológico Clara Campal and START Madrid, Madrid, Spain
| | - Camillio Porta
- Medical Oncology, I.R.C.C.S. San Matteo University Hospital Foundation, Pavia, Italy
| | - Viktor Grünwald
- Clinic for Hematology, Hemostaseology, Oncology & Stem Cell Transplantation, Medical School of Hannover, Hannover, Germany
| | - Bernard Escudier
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| |
Collapse
|
141
|
Sola AM, Johnson DE, Grandis JR. Investigational multitargeted kinase inhibitors in development for head and neck neoplasms. Expert Opin Investig Drugs 2019; 28:351-363. [PMID: 30753792 DOI: 10.1080/13543784.2019.1581172] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Despite advances in treatment, head and neck squamous cell carcinoma (HNSCC) survival rates remain stagnant. Current treatment is associated with significant toxicities and includes chemotherapy, radiation, surgery, and few targeted treatments. Targeted treatments, epidermal growth factor receptor (EGFR)-targeted agent, cetuximab, and immune checkpoint inhibitors, pembrolizumab and nivolumab, show improved toxicity profiles and modestly improved survival in select patients. An urgent need remains to identify novel targeted treatments for single-agent or combined therapy use. AREAS COVERED Multitargeted kinase inhibitors are small molecule inhibitors with limited toxicity. This review will focus on early-stage investigations of multitargeted tyrosine kinase inhibitors (m-TKIs) (those that target at least two tyrosine kinases) for HNSCC. Preclinical and early trials investigating m-TKIs for various disease settings of HNSCC will be evaluated for efficacy, identification of significant biomarkers and potential for combination therapy. EXPERT OPINION Few single agent m-TKIs have demonstrated efficacy in unselected HNSCC populations. The most promising clinical results have been obtained when m-TKIs are tested in combination with other therapies, including immunotherapy, or in mutation-defined subgroups of patients. The future success of m-TKIs will rely on identification, in preclinical models and clinical trials, of predictive biomarkers of response and mechanisms of innate and acquired resistance.
Collapse
Affiliation(s)
- Ana Marija Sola
- a Department of Otolaryngology - Head and Neck Surgery , University of California , San Francisco , CA , USA
| | - Daniel E Johnson
- a Department of Otolaryngology - Head and Neck Surgery , University of California , San Francisco , CA , USA
| | - Jennifer R Grandis
- a Department of Otolaryngology - Head and Neck Surgery , University of California , San Francisco , CA , USA
| |
Collapse
|
142
|
Abstract
Drug-induced pigmentation accounts for up to 20% of all cases of acquired pigmentation. A thorough review of medical history and previous and ongoing medications as well as a complete skin examination can guide diagnosis. Implicated agents include alkylating/cytotoxic agents, analgesics, antiarrhythmics, anticoagulants, antiepileptics, antimalarials, antimicrobials, antiretrovirals, metals, prostaglandin analogs, and psychotropic agents, among others. Confirming true drug associations can be challenging, especially in the setting of delayed onset of pigmentation and coexisting polypharmacy.
Collapse
Affiliation(s)
- Amanda F Nahhas
- Department of Dermatology, Beaumont-Farmington Hills, Farmington Hills, MI, USA
- Department of Dermatology, Henry Ford Hospital, 3031 W. Grand Blvd., Suite 800, Detroit, MI, 48202, USA
| | - Taylor L Braunberger
- Department of Dermatology, Henry Ford Hospital, 3031 W. Grand Blvd., Suite 800, Detroit, MI, 48202, USA
| | - Iltefat H Hamzavi
- Department of Dermatology, Henry Ford Hospital, 3031 W. Grand Blvd., Suite 800, Detroit, MI, 48202, USA.
| |
Collapse
|
143
|
Chatziathanasiadou MV, Stylos EK, Giannopoulou E, Spyridaki MH, Briasoulis E, Kalofonos HP, Crook T, Syed N, Sivolapenko GB, Tzakos AG. Development of a validated LC-MS/MS method for the in vitro and in vivo quantitation of sunitinib in glioblastoma cells and cancer patients. J Pharm Biomed Anal 2019; 164:690-697. [DOI: 10.1016/j.jpba.2018.11.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 11/05/2018] [Accepted: 11/12/2018] [Indexed: 12/28/2022]
|
144
|
Kim JH, Ryu MH, Yoo C, Chae H, Na H, Beck M, Kim BS, Yoo MW, Yook JH, Kim BS, Kim KH, Kim CW, Kang YK. Long-term survival outcome with tyrosine kinase inhibitors and surgical intervention in patients with metastatic or recurrent gastrointestinal stromal tumors: A 14-year, single-center experience. Cancer Med 2019; 8:1034-1043. [PMID: 30693663 PMCID: PMC6434201 DOI: 10.1002/cam4.1994] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/11/2018] [Accepted: 01/08/2019] [Indexed: 12/17/2022] Open
Abstract
The long‐term effects of tyrosine kinase inhibitors (TKIs), including imatinib, and surgical intervention on advanced gastrointestinal stromal tumor (GIST) were evaluated. All 379 patients had metastatic or recurrent GIST and started 400 mg/d imatinib at the Asan Medical Center in periods 1 and 2 [2001‐2007 (33.2%) and 2008‐2014 (66.8%), respectively]. Men constituted 60.4%; median patient age and tumor size at the initiation of imatinib were 58.6 (14.6‐85.5) years and 51 (0‐324) mm, respectively, without differences between periods except for older age and less preimatinib surgery in period 2. Response and disease control rates with imatinib in measurable GIST were 63.1% and 94.3%, respectively, without differences between periods. More patients in period 2 underwent surgical resection for TKI‐responsive diseases within the first 2 years (24.9%, P = 0.006). With a median follow‐up of 6.1 years (2.5‐16.0) in survivors, median progression‐free survival (PFS) was 5.4 years [95% confidence interval (CI), 4.0‐6.9]. Subsequent sunitinib (P = 0.066) and regorafenib (P = 0.003) were more commonly administered in period 2. Median overall survival (OS) was 8.8 years (95% CI, 7.8‐9.7). PFS with imatinib (P = 0.002) and OS (P = 0.019) were significantly longer in period 2. Young age, smaller tumor size at the initiation of imatinib, KIT exon 11 mutation, surgical intervention, and period 2 were favorable factors for PFS and OS. Patients with advanced GIST showed better prognosis with the optimal use of imatinib, along with active surgical intervention and more common use of subsequent TKIs in period 2.
Collapse
Affiliation(s)
- Jwa Hoon Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Min-Hee Ryu
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Changhoon Yoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Heejung Chae
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hana Na
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Moyoul Beck
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Beom Su Kim
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Moon-Won Yoo
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jeong Hwan Yook
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Byung Sik Kim
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ki-Hun Kim
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Chan Wook Kim
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Yoon-Koo Kang
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
145
|
Chandra F, Zaks L, Zhu A. Survival Prolongation Index as a Novel Metric to Assess Anti-Tumor Activity in Xenograft Models. AAPS JOURNAL 2019; 21:16. [PMID: 30627814 DOI: 10.1208/s12248-018-0284-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 12/11/2018] [Indexed: 12/15/2022]
Abstract
A single efficacy metric quantifying anti-tumor activity in xenograft models is useful in evaluating different tumors' drug sensitivity and dose-response of an anti-tumor agent. Commonly used metrics include the ratio of tumor volume in treated vs. control mice (T/C), tumor growth inhibition (TGI), ratio of area under the curve (AUC), and growth rate inhibition (GRI). However, these metrics have some limitations. In particular, for biologics with long half-lives, tumor volume (TV) of treated xenografts displays a delay in volume reduction (and in some cases, complete regression) followed by a growth rebound. These observed data cannot be described by exponential functions, which is the underlying assumption of TGI and GRI, and the fit depends on how long the tumor volumes are monitored. On the other hand, T/C and TGI only utilizes information from one chosen time point. Here, we propose a new metric called Survival Prolongation Index (SPI), calculated as the time for drug-treated TV to reach a certain size (e.g., 600 mm3) divided by the time for control TV to reach 600mm3 and therefore not dependent on the chosen final time point tf. Simulations were conducted under different scenarios (i.e., exponential vs. saturable growth, linear vs. nonlinear kill function). For all cases, SPI is the most linear and growth-rate independent metric. Subsequently, a literature analysis was conducted using 11 drugs to evaluate the correlation between pre-clinically obtained SPI and clinical overall response. This retrospective analysis of approved drugs suggests that a predicted SPI of 2 is necessary for clinical response.
Collapse
Affiliation(s)
- Fiona Chandra
- Translation Modeling and Simulation, DMPK, Takeda Pharmaceuticals, 35 Landsdowne St, Cambridge, Massachusetts, 02139, USA.
| | - Lihi Zaks
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Andy Zhu
- Translation Modeling and Simulation, DMPK, Takeda Pharmaceuticals, 35 Landsdowne St, Cambridge, Massachusetts, 02139, USA
| |
Collapse
|
146
|
Rovithi M, Gerritse SL, Honeywell RJ, Ten Tije AJ, Ruijter R, Peters GJ, Voortman J, Labots M, Verheul HMW. Phase I Dose-Escalation Study of Once Weekly or Once Every Two Weeks Administration of High-Dose Sunitinib in Patients With Refractory Solid Tumors. J Clin Oncol 2018; 37:411-418. [PMID: 30586316 PMCID: PMC6368417 DOI: 10.1200/jco.18.00725] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
PURPOSE Dose and schedule optimization of treatment with tyrosine kinase inhibitors is of utmost importance. On the basis of preclinical data, a phase I clinical trial of once weekly or once every 2 weeks administration of high-dose sunitinib in patients with refractory solid malignancies was conducted. PATIENTS AND METHODS Patients with advanced cancer refractory to standard treatment were eligible. With use of a standard 3 + 3 phase I design, patients received escalating doses of sunitinib, in 100 mg increments, starting at 200 mg once weekly. In both the once weekly and once every 2 weeks cohorts, 10 more patients were included at the maximum tolerated dose level. Primary end points were safety and tolerability. RESULTS Sixty-nine patients with advanced cancer, predominantly colorectal cancer (42%), were treated with this alternative dosing regimen. Maximum tolerated dose was established at 300 mg once weekly and 700 mg once every 2 weeks, resulting in nine- and 18-fold higher maximum plasma concentrations compared with standard dose, respectively. Treatment was well tolerated, with fatigue (81%), nausea (48%), and anorexia (33%) being the most frequent adverse events. The only grade 3 or 4 treatment-related adverse event in 5% or more of patients was fatigue (6%). Sixty-three percent of patients had significant clinical benefit, with a 30% progression-free survival of 5 months or more. CONCLUSION Sunitinib administered once weekly at 300 mg or once every 2 weeks at 700 mg is feasible, with comparable tolerability as daily administration. Administration of 700 mg once every 2 weeks can be considered as the most optimal schedule because of the highest maximum plasma concentration being reached. The promising preliminary antitumor activity of this alternative schedule in heavily pretreated patients warrants further clinical evaluation and might ultimately indicate a class characteristic of tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Maria Rovithi
- Vrije Universiteit Medical Center, Amsterdam, the Netherlands
| | | | | | | | - Rita Ruijter
- Vrije Universiteit Medical Center, Amsterdam, the Netherlands
| | | | - Jens Voortman
- Vrije Universiteit Medical Center, Amsterdam, the Netherlands
| | - Mariette Labots
- Vrije Universiteit Medical Center, Amsterdam, the Netherlands
| | | |
Collapse
|
147
|
Ahmad S, Singh V, Sinha R, Srivastava A, Mandhani A. Role of MMP-2, MMP-9 and VEGF as serum biomarker in early prognosis of renal cell carcinoma. AFRICAN JOURNAL OF UROLOGY 2018. [DOI: 10.1016/j.afju.2018.09.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
148
|
Bendell JC, Patel MR, Moore KN, Chua CC, Arkenau HT, Dukart G, Harrow K, Liang C. Phase I, First-in-Human, Dose-Escalation Study to Evaluate the Safety, Tolerability, and Pharmacokinetics of Vorolanib in Patients with Advanced Solid Tumors. Oncologist 2018; 24:455-e121. [PMID: 30478190 PMCID: PMC6459237 DOI: 10.1634/theoncologist.2018-0740] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 10/28/2018] [Indexed: 11/17/2022] Open
Abstract
Lessons Learned. Pharmacokinetic results underscore that the vorolanib (X‐82) study design was successful without the need for further dose escalation beyond 400 mg once daily (q.d.). Therefore, the recommended dose of X‐82 as a single agent in patients with advanced cancer is 400 mg q.d.
Background. Vorolanib (X‐82) is a novel, oral, multikinase vascular endothelial growth factor (VEGF) receptor/platelet‐derived growth factor (PDGF) receptor inhibitor that was developed on the same chemical scaffold as sunitinib, but designed to improve upon the safety profile while maintaining the efficacy of sunitinib. By targeting the VEGF and PDGF receptors, X‐82 was expected to disrupt tumor angiogenesis and be active in a broad spectrum of solid tumors. Therefore, we determined the maximum tolerated dose (MTD) and characterized the preliminary pharmacokinetics and clinical tumor response of X‐82 as a single agent in patients with advanced solid tumors. Methods. Adult patients with advanced solid tumors received X‐82 as tablets or capsules (once daily [q.d.] or b.i.d.) every 4 weeks. Patients were evaluated for response every 8 weeks, and continued treatment until disease progression or intolerable toxicity. Results. Fifty‐two patients received study treatment in 17 cohorts. X‐82 capsule dosing was as follows: cohorts 1–6 (20–400 mg q.d.) and cohorts 7–8 (140–200 mg b.i.d.). Patients in cohorts 9–17 received 50–800 mg q.d. tablet dosing. The median time on treatment was 58 days. X‐82 blood pharmacokinetics appeared dose‐independent with a t1/2 of 5.13 hours and 6.48 hours for capsule and tablet formulations, respectively. No apparent accumulation was observed after 21 days of daily dosing. Conclusion. X‐82 had a safety profile consistent with its mechanism of action. It has a short half‐life and was well tolerated by most patients. Study enrollment ended prior to the determination of the MTD because of the apparent saturation of absorption at 400–800 mg. The recommended dose of X‐82 as a single agent in patients with advanced cancer is 400 mg q.d.
Collapse
Affiliation(s)
- Johanna C Bendell
- Sarah Cannon Research Institute, Nashville, Tennessee, USA
- Tennessee Oncology, PLLC, Nashville, Tennessee, USA
| | - Manish R Patel
- Sarah Cannon Research Institute, Nashville, Tennessee, USA
- Florida Cancer Specialists & Research Institute, Sarasota, Florida, USA
| | - Kathleen N Moore
- Sarah Cannon Research Institute, Nashville, Tennessee, USA
- Peggy and Charles Stephenson Oklahoma Cancer Center, Oklahoma City, Oklahoma, USA
| | | | | | - Gary Dukart
- Equinox Sciences, LLC, Palm Beach Gardens, Florida, USA
| | - Kim Harrow
- Equinox Sciences, LLC, Palm Beach Gardens, Florida, USA
| | - Chris Liang
- Equinox Sciences, LLC, Palm Beach Gardens, Florida, USA
| |
Collapse
|
149
|
Tsuchiya N. Molecular-targeted therapy in advanced renal cell carcinoma based on pharmacokinetics, pharmacodynamics and pharmacogenetics: A proposed strategy. Int J Urol 2018; 26:48-56. [DOI: 10.1111/iju.13805] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 08/20/2018] [Indexed: 01/16/2023]
Affiliation(s)
- Norihiko Tsuchiya
- Department of Urology; Yamagata University Faculty of Medicine; Yamagata Japan
| |
Collapse
|
150
|
Zhang X, Sun G, Zhao J, Shu K, Zhao P, Liu J, Yang Y, Tang Q, Chen J, Shen P, Wang J, Zeng H. Improved Long-Term Clinical Outcomes And Safety Profile Of Sunitinib Dosing Schedule With 4/2 Switched To 2/1 In Patients With Metastatic Renal Cell Carcinoma. J Cancer 2018; 9:3303-3310. [PMID: 30271490 PMCID: PMC6160671 DOI: 10.7150/jca.25693] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 06/16/2018] [Indexed: 02/05/2023] Open
Abstract
Purpose: This study aimed to identify the survival benefit and safety of alternative dosage schedules for sunitinib in metastatic renal cell carcinoma. Materials and Methods: Clinicopathologic and survival data of patients treated with sunitinib as first-line therapy were retrospectively reviewed. Patients were classified into three groups: a standard dosing schedule (4/2 schedule), alternative dosing schedule (2/1 schedule), and switched dosing schedule (4/2-2/1 schedule). Results: Ninety-nine patients were retrospectively included. Seventy-five (75.8%) patients were initially administrated with a 4/2 schedule of sunitinib, while 24 were started with the 2/1 schedule. During treatment, 45 (60.0%) patients with an initial 4/2 schedule switched to a 2/1 schedule (4/2-2/1 schedule) due to severe adverse events (AEs) or poor tolerance. Compared to that with a 4/2 schedule, patients with a 2/1 schedule had a much lower incidence of grade 3/4 AEs (69.6% vs. 40.6%, p=0.001). Overall, the 4/2-2/1 schedule was associated with the best survival benefits. Among the 4/2, 2/1, and 4/2-2/1 schedule groups, the median PFS was 12.5, 11.0, and 25.0 months, respectively (p=0.003), and the median OS was 21.0, 28.0, and 52.0 months, respectively (p=0.03). Multivariate analysis identified the 4/2-2/1 schedule as an independent factor predicting favorable PFS. Although without statistical significance, 4/2-2/1 schedule could decrease 55% risk of death. Furthermore, patients with unfavorable IMDC risk seemed to have more opportunity to achieve better survival from the 4/2-2/1 dosing schedule. Conclusion: Patients with a 4/2-2/1 schedule could minimize treatment-related toxicities; more importantly, patients with 4/2-2/1 schedule could achieve a superior survival benefit.
Collapse
Affiliation(s)
- Xingming Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China, 610041
| | - Guangxi Sun
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China, 610041
| | - Jinge Zhao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China, 610041
| | - Kunpeng Shu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China, 610041
| | - Peng Zhao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China, 610041
| | - Jiandong Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China, 610041
| | - Yaojing Yang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China, 610041
| | - Qidun Tang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China, 610041
| | - Junru Chen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China, 610041
| | - Pengfei Shen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China, 610041
| | - Jia Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China, 610041
| | - Hao Zeng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China, 610041
| |
Collapse
|