1
|
Gong J, Zhang W, Balthasar JP. Camptothein-Based Anti-Cancer Therapies and Strategies to Improve Their Therapeutic Index. Cancers (Basel) 2025; 17:1032. [PMID: 40149365 PMCID: PMC11941615 DOI: 10.3390/cancers17061032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/06/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
Camptothecin and its derivatives (CPTs) are potent antineoplastic agents that exert their effects by inhibiting DNA topoisomerase I, leading to apoptosis during cell proliferation. Since their discovery in the 1960s, CPTs have faced challenges such as low water solubility, pH-dependent lactone ring instability, and severe off-target toxicities. Despite extensive research, only two CPTs, irinotecan and topotecan, have received health authority approval. Ongoing clinical trials continue to explore the use of CPTs in combination with targeted therapies and immunotherapies to expand their clinical use. Drug delivery systems, including liposomes and antibody-drug conjugates (ADCs), have significantly enhanced the therapeutic index of CPTs. Liposomal irinotecan (Onivyde®, Ipsen, Paris, France) and two ADCs delivering CPT payloads, trastuzumab deruxtecan (Enhertu®, Daiichi Sankyo, Tokyo, Japan) and sacituzumab govitecan (Trodelvy®, Gilead Sciences, Inc., Foster City, CA, USA), have demonstrated substantial efficacy and safety. There is promise that novel strategies such as inverse targeting and co-dosing with anti-idiotypic distribution enhancers may expand the utility of CPT ADCs. This review highlights CPT therapies in clinical use and discusses approaches to further enhance their therapeutic selectivity.
Collapse
Affiliation(s)
| | | | - Joseph P. Balthasar
- Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, NY 14214, USA (W.Z.)
| |
Collapse
|
2
|
Zarnoosheh Farahani T, Nejadmoghaddam MR, Sari S, Ghahremanzadeh R, Minai-Tehrani A. Generation of anti-SN38 antibody for loading efficacy and therapeutic monitoring of SN38-containing therapeutics. Heliyon 2024; 10:e33232. [PMID: 39021912 PMCID: PMC11253049 DOI: 10.1016/j.heliyon.2024.e33232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 06/17/2024] [Accepted: 06/17/2024] [Indexed: 07/20/2024] Open
Abstract
SN38, one of the most potent anti-tumor analogues of the camptothecins (CPTs), has limitations in its direct formulation as an anticancer agent due to its super toxicity and poor solubility in water and pharmaceutically approved solvents. However, it has garnered significant scientific interest as a payload in conjugated nanomedicine platforms (e.g., SN-38lip, NK012, SNB-101, and ADCs) to enhance their effectiveness and safety. The development of these platforms necessitates a convenient quantitative determination of SN38 in preclinical and clinical studies, a need that our study directly addresses, offering a practical solution to a pressing problem in cancer research and drug development. This study details the meticulous process of generating poly and monoclonal antibodies (pAb and mAb) against SN38 and their application to measure the SN38 in naked and conjugated forms of SN38-conjugated ADCs. For this purpose, two haptens of SN38 were synthesized by introducing the glycine or 4-amino-4-oxobutanyol(glycine) moiety as a conjugation functional group of the SN38. IR, NMR and mass spectrometric techniques confirmed the chemical modifications of the haptens. The haptens were then conjugated to each bovine serum albumin (BSA) and keyhole limpet hemocyanin (KLH) protein. The SN38-KLH conjugates were meticulously examined for immunization and generation of pAb and mAb. The immunization efficiency, reactivity, binding affinity, specificity, and cross-reactivity of purified pAb and mAb against Irinotecan, a model for the emergence of an SN38 derivative in clinical settings, were evaluated using ELISA and western blotting (WB) techniques. Conjugation efficiency of the SN38 to the KLH was increased using 4-amino-4-oxobutanyol(glycine) moiety, as its immunization efficacy was more to generate pAb. Furthermore, only this hapten could immunized mice to generate mAb recognizing SN38 with nanomolar equilibrium affinity. Our recent findings strongly support the notion that the generated pAb employed in developing an ELISA effectively ascertains the presence of SN38 in SN38-conjugated ADC, with a test midpoint EC50 of 2.5 μg/mL. Our study's unique contribution to the field lies in the development of specific antibodies against SN38 for measuring it on ADC, a feat that has not been achieved before. These immunoassays can be readily applied to detect other SN38-conjugate therapeutic platforms, thereby enhancing their clinical knowledge translation. The affinity of both pAb and mAb also meets the acceptance criteria for quantifying SN38 in fluidic material, as well as in Therapeutic drug monitoring (TDM) studies, a crucial aspect of personalized medicine. The potential applications of the anti-SN38 antibodies extend to reducing SN38-induced systemic toxicity through an inverse targeting strategy, a novel approach that piques further interest in our findings.
Collapse
Affiliation(s)
- Tahereh Zarnoosheh Farahani
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | | | - Soyar Sari
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ramin Ghahremanzadeh
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Arash Minai-Tehrani
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| |
Collapse
|
3
|
Yu C, Huang F, Wang K, Liu M, Chow WA, Ling X, Li F, Causey JL, Huang X, Cook-Wiens G, Cui X. Single protein encapsulated SN38 for tumor-targeting treatment. J Transl Med 2023; 21:897. [PMID: 38072965 PMCID: PMC10712105 DOI: 10.1186/s12967-023-04778-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND The alkaloid camptothecin analog SN38 is a potent antineoplastic agent, but cannot be used directly for clinical application due to its poor water solubility. Currently, the prodrug approach on SN38 has resulted in 3 FDA-approved cancer therapeutics, irinotecan, ONIVYDE, and Trodelvy. However, only 2-8% of irinotecan can be transformed enzymatically in vivo into the active metabolite SN38, which severely limits the drug's efficacy. While numerous drug delivery systems have been attempted to achieve effective SN38 delivery, none have produced drug products with antitumor efficacy better than irinotecan in clinical trials. Therefore, novel approaches are urgently needed for effectively delivering SN38 to cancer cells with better efficacy and lower toxicity. METHODS Based on the unique properties of human serum albumin (HSA), we have developed a novel single protein encapsulation (SPE) technology to formulate cancer therapeutics for improving their pharmacokinetics (PK) and antitumor efficacy and reducing their side effects. Previous application of SPE technology to doxorubicin (DOX) formulation has led to a promising drug candidate SPEDOX-6 (FDA IND #, 152154), which will undergo a human phase I clinical trial. Using the same SPE platform on SN38, we have now produced two SPESN38 complexes, SPESN38-5 and SPESN38-8. We conducted their pharmacological evaluations with respect to maximum tolerated dose, PK, and in vivo efficacy against colorectal cancer (CRC) and soft tissue sarcoma (STS) in mouse models. RESULTS The lyophilized SPESN38 complexes can dissolve in aqueous media to form clear and stable solutions. Maximum tolerated dose (MTD) of SPESN38-5 is 250 mg/kg by oral route (PO) and 55 mg/kg by intravenous route (IV) in CD-1 mice. SPESN38-8 has the MTD of 45 mg/kg by IV in the same mouse model. PK of SPESN38-5 by PO at 250 mg/kg gave mouse plasma AUC0-∞ of 0.05 and 4.5 nmol × h/mL for SN38 and SN38 glucuronidate (SN38G), respectively, with a surprisingly high molar ratio of SN38G:SN38 = 90:1. However, PK of SPESN38-5 by IV at 55 mg/kg yielded much higher mouse plasma AUC0-∞ of 19 and 28 nmol × h/mL for SN38 and SN38G, producing a much lower molar ratio of SN38G:SN38 = 1.5:1. Antitumor efficacy of SPESN38-5 and irinotecan (control) was evaluated against HCT-116 CRC xenograft tumors. The data indicates that SPESN38-5 by IV at 55 mg/kg is more effective in suppressing HCT-116 tumor growth with lower systemic toxicity compared to irinotecan at 50 mg/kg. Additionally, SPESN38-8 and DOX (control) by IV were evaluated in the SK-LMS-1 STS mouse model. The results show that SPESN38-8 at 33 mg/kg is highly effective for inhibiting SK-LMS-1 tumor growth with low toxicity, in contrast to DOX's insensitivity to SK-LMS-1 with high toxicity. CONCLUSION SPESN38 complexes provide a water soluble SN38 formulation. SPESN38-5 and SPESN38-8 demonstrate better PK values, lower toxicity, and superior antitumor efficacy in mouse models, compared with irinotecan and DOX.
Collapse
Affiliation(s)
- Changjun Yu
- Department of Chemistry, California Institute of Technology, Pasadena, CA, 91125, USA.
- Sunstate Biosciences, LLC, 870 S. Myrtle Ave, Monrovia, CA, 91016, USA.
| | - Faqing Huang
- Department of Chemistry and Biochemistry, University of Southern Mississippi, Hattiesburg, MS, 39406, USA.
| | - Kinsley Wang
- Sunstate Biosciences, LLC, 870 S. Myrtle Ave, Monrovia, CA, 91016, USA
| | - Mengmeng Liu
- Sunstate Biosciences, LLC, 870 S. Myrtle Ave, Monrovia, CA, 91016, USA
| | - Warren A Chow
- Division of Hematology/Oncology, Department of Medicine, UCI Health, Orange, CA, 92868, USA
| | - Xiang Ling
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
- Canget BioTekpharma, LLC, 701 Ellicott Street, Buffalo, NY, 14203, USA
| | - Fengzhi Li
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Jason L Causey
- Department of Computer Sciences, Arkansas State University, Jonesboro, AR, 72467, USA
| | - Xiuzhen Huang
- Department of Computational Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Galen Cook-Wiens
- Department of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Xiaojiang Cui
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars Sinai Medical Center, Los Angeles, CA, 90048, USA.
| |
Collapse
|
4
|
Yu CJ, Huang F, Wang K, Liu M, Chow WA, Ling X, Li F, Causey JL, Huang X, Cook-Wiens G, Cui X. Single Protein Encapsulated SN38 for Tumor-Targeting Treatment. RESEARCH SQUARE 2023:rs.3.rs-3154635. [PMID: 37546894 PMCID: PMC10402254 DOI: 10.21203/rs.3.rs-3154635/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Background The alkaloid camptothecin analog SN38 is a potent antineoplastic agent, but cannot be used directly for clinical application due to its poor water solubility. Currently, the prodrug approach on SN38 has resulted in 3 FDA-approved cancer therapeutics, irinotecan, ONIVYDE, and Trodelvy. However, only 2-8% of irinotecan can be transformed enzymatically in vivo into the active metabolite SN38, which severely limits the drug's efficacy. While numerous drug delivery systems have been attempted to achieve effective SN38 delivery, none have produced drug products with antitumor efficacy better than irinotecan in clinical trials. Therefore, novel approaches are urgently needed for effectively delivering SN38 to cancer cells with better efficacy and lower toxicity. Methods Based on the unique properties of human serum albumin (HSA), we have developed a novel single protein encapsulation (SPE) technology to formulate cancer therapeutics for improving their pharmacokinetics (PK) and antitumor efficacy and reducing their side effects. Previous application of SPE technology to doxorubicin (DOX) formulation has led to a promising drug candidate SPEDOX-6 (FDA IND #, 152154), which will undergo a human phase I clinical trial. Using the same SPE platform on SN38, we have now produced two SPESN38 complexes, SPESN38-5 and SPESN38-8. We conducted their pharmacological evaluations with respect to maximum tolerated dose, PK, and in vivo efficacy against colorectal cancer (CRC) and soft tissue sarcoma (STS) in mouse models. Results The lyophilized SPESN38 complexes can dissolve in aqueous media to form clear and stable solutions. Maximum tolerated dose (MTD) of SPESN38-5 is 250 mg/kg by oral route (PO) and 55 mg/kg by intravenous route (IV) in CD-1 mice. SPESN38-8 has the MTD of 45 mg/kg by IV in the same mouse model. PK of SPESN38-5 by PO at 250 mg/kg gave mouse plasma AUC0-∞ of 0.0548 and 4.5007 (nmol × h/mL) for SN38 and SN38 glucuronidate (SN38G), respectively, with a surprisingly high molar ratio of SN38G:SN38 = 82:1. However, PK of SPESN38-5 by IV at 55 mg/kg yielded much higher mouse plasma AUC0-∞ of 18.80 and 27.78 nmol × h/mL for SN38 and SN38G, producing a much lower molar ratio of SN38G:SN38 = 1.48:1. Antitumor efficacy of SPESN38-5 and irinotecan (control) was evaluated against HCT-116 CRC xenograft tumors. The data indicates that SPESN38-5 by IV at 55 mg/kg is more effective in suppressing HCT-116 tumor growth with lower systemic toxicity compared to irinotecan at 50 mg/kg. Additionally, SPESN38-8 and DOX (control) by IV were evaluated in the SK-LMS-1 STS mouse model. The results show that SPESN38-8 at 33 mg/kg is highly effective for inhibiting SK-LMS-1 tumor growth with low toxicity, in contrast to DOX's insensitivity to SK-LMS-1 with high toxicity. Conclusion SPESN38 complexes provide a water soluble SN38 formulation. SPESN38-5 and SPESN38-8 demonstrate better PK values, lower toxicity, and superior antitumor efficacy in mouse models, compared with irinotecan and DOX.
Collapse
Affiliation(s)
| | - Faqing Huang
- University of Southern Mississippi Center For Tobacco Prevention and Health Promotion: University of Southern Mississippi
| | | | | | - Warren A Chow
- University of California Irvine Department of Medicine
| | - Xiang Ling
- Roswell Park Comprehensive Cancer Center
| | - Fengzhi Li
- Roswell Park Comprehensive Cancer Center
| | | | | | | | | |
Collapse
|
5
|
Lopez-Mendez TB, Strippoli R, Trionfetti F, Calvo P, Cordani M, Gonzalez-Valdivieso J. Clinical Trials Involving Chemotherapy-Based Nanocarriers in Cancer Therapy: State of the Art and Future Directions. Cancer Nanotechnol 2023. [DOI: 10.1007/978-3-031-17831-3_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
6
|
Gholami L, Ivari JR, Nasab NK, Oskuee RK, Sathyapalan T, Sahebkar A. Recent Advances in Lung Cancer Therapy Based on Nanomaterials: A Review. Curr Med Chem 2023; 30:335-355. [PMID: 34375182 DOI: 10.2174/0929867328666210810160901] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/22/2021] [Accepted: 06/26/2021] [Indexed: 11/22/2022]
Abstract
Lung cancer is one of the commonest cancers with a significant mortality rate for both genders, particularly in men. Lung cancer is recognized as one of the leading causes of death worldwide, which threatens the lives of over 1.6 million people every day. Although cancer is the leading cause of death in industrialized countries, conventional anticancer medications are unlikely to increase patients' life expectancy and quality of life significantly. In recent years, there are significant advances in the development and applications of nanotechnology in cancer treatment. The superiority of nanostructured approaches is that they act more selectively than traditional agents. This progress led to the development of a novel field of cancer treatment known as nanomedicine. Various formulations based on nanocarriers, including lipids, polymers, liposomes, nanoparticles and dendrimers have opened new horizons in lung cancer therapy. The application and expansion of nano-agents lead to an exciting and challenging research era in pharmaceutical science, especially for the delivery of emerging anti-cancer agents. The objective of this review is to discuss the recent advances in three types of nanoparticle formulations for lung cancer treatments modalities, including liposomes, polymeric micelles, and dendrimers for efficient drug delivery. Afterward, we have summarized the promising clinical data on nanomaterials based therapeutic approaches in ongoing clinical studies.
Collapse
Affiliation(s)
- Leila Gholami
- Nanotechnology Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jalil Rouhani Ivari
- Department of Medical Biotechnology and Nanotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Niloofar Khandan Nasab
- Department of Medical Biotechnology and Nanotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Kazemi Oskuee
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Thozhukat Sathyapalan
- Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, United Kingdom of Great Britain and Northern Ireland
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran.,School of Medicine, The University of Western Australia, Perth, Australia.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
7
|
Emerging concepts in designing next-generation multifunctional nanomedicine for cancer treatment. Biosci Rep 2022; 42:231373. [PMID: 35638450 PMCID: PMC9272595 DOI: 10.1042/bsr20212051] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 05/10/2022] [Accepted: 05/12/2022] [Indexed: 11/17/2022] Open
Abstract
Nanotherapy has emerged as an improved anticancer therapeutic strategy to circumvent the harmful side effects of chemotherapy. It has been proven to be beneficial to offer multiple advantages, including their capacity to carry different therapeutic agents, longer circulation time and increased therapeutic index with reduced toxicity. Over time, nanotherapy evolved in terms of their designing strategies like geometry, size, composition or chemistry to circumvent the biological barriers. Multifunctional nanoscale materials are widely used as molecular transporter for delivering therapeutics and imaging agents. Nanomedicine involving multi-component chemotherapeutic drug-based combination therapy has been found to be an improved promising approach to increase the efficacy of cancer treatment. Next-generation nanomedicine has also utilized and combined immunotherapy to increase its therapeutic efficacy. It helps in targeting tumor immune response sparing the healthy systemic immune function. In this review, we have summarized the progress of nanotechnology in terms of nanoparticle designing and targeting cancer. We have also discussed its further applications in combination therapy and cancer immunotherapy. Integrating patient-specific proteomics and biomarker based information and harnessing clinically safe nanotechnology, the development of precision nanomedicine could revolutionize the effective cancer therapy.
Collapse
|
8
|
Mi P, Miyata K, Kataoka K, Cabral H. Clinical Translation of Self‐Assembled Cancer Nanomedicines. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000159] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Peng Mi
- Department of Radiology, Center for Medical Imaging, State Key Laboratory of Biotherapy and Cancer Center West China Hospital, Sichuan University No. 17 People's South Road Chengdu 610041 China
| | - Kanjiro Miyata
- Department of Materials Engineering, Graduate School of Engineering The University of Tokyo 7‐3‐1 Hongo, Bunkyo‐ku Tokyo 113‐8656 Japan
| | - Kazunori Kataoka
- Institute for Future Initiatives The University of Tokyo 7‐3‐1 Hongo, Bunkyo‐ku Tokyo 113‐0033 Japan
- Innovation Center of NanoMedicine Kawasaki Institute of Industrial Promotion 3‐25‐14, Tonomachi, Kawasaki‐ku Kawasaki 210‐0821 Japan
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering The University of Tokyo 7‐3‐1 Hongo, Bunkyo‐ku Tokyo 113‐8656 Japan
| |
Collapse
|
9
|
Rodallec A, Benzekry S, Lacarelle B, Ciccolini J, Fanciullino R. Pharmacokinetics variability: Why nanoparticles are not just magic-bullets in oncology. Crit Rev Oncol Hematol 2018; 129:1-12. [PMID: 30097227 DOI: 10.1016/j.critrevonc.2018.06.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/16/2018] [Accepted: 06/13/2018] [Indexed: 12/11/2022] Open
Abstract
Developing nanoparticles to improve the specificity of anticancer agents towards tumor tissue and to better control drug delivery is a rising strategy in oncology. An increasing number of forms (e.g., conjugated nanoparticles, liposomes, immunoliposomes…) are now available on the shelves and numerous other scaffolds (e.g., dendrimeres, nanospheres, squalenes …) are currently at various stages of development. However, as of today most nanoparticles made available remain lipidic carriers. Pharmacokinetic variability is a major, yet largely underestimated issue with liposomal nanoparticles. A wide variety of causes (e.g., tumor type and disease staging, comorbidities, patient's immune system) can explain this variability, which can in return negatively impact pharmacodynamic endpoints such as poor efficacy or severe toxicities. This review aims to cover the main causes for erratic pharmacokinetics observed with most nanoparticles, especially liposomes used in oncology. Should the main causes of such variability be identified, specific studies in non-clinical or clinical development stages could be undertaken using dedicated models (i.e., mechanistic or semi-mechanistic mathematical models such as PBPK approaches) to better describe nanoparticles pharmacokinetics and decipher PK/PD relationships. In addition, identifying relevant biomarkers or parameters likely to impact nanoparticles pharmacokinetics would allow for either the modification of their characteristics to reduce the influence of the expected variability during development phases or the development of biomarker-based adaptive dosing strategies to maintain an optimal efficacy/toxicity balance. Broadly, we call for the development of comprehensive distribution studies and state-of-the-art modeling support to better understand and anticipate nanoparticle pharmacokinetics in oncology.
Collapse
Affiliation(s)
- Anne Rodallec
- SMARTc unit, Center for Research on Cancer of Marseille (CRCM): UMR Inserm 1068, CNRS UMR 7258, Aix Marseille Université, Marseille, France
| | | | - Bruno Lacarelle
- SMARTc unit, Center for Research on Cancer of Marseille (CRCM): UMR Inserm 1068, CNRS UMR 7258, Aix Marseille Université, Marseille, France
| | - Joseph Ciccolini
- SMARTc unit, Center for Research on Cancer of Marseille (CRCM): UMR Inserm 1068, CNRS UMR 7258, Aix Marseille Université, Marseille, France
| | - Raphaelle Fanciullino
- SMARTc unit, Center for Research on Cancer of Marseille (CRCM): UMR Inserm 1068, CNRS UMR 7258, Aix Marseille Université, Marseille, France.
| |
Collapse
|
10
|
Chang TC, Shiah HS, Yang CH, Yeh KH, Cheng AL, Shen BN, Wang YW, Yeh CG, Chiang NJ, Chang JY, Chen LT. Phase I study of nanoliposomal irinotecan (PEP02) in advanced solid tumor patients. Cancer Chemother Pharmacol 2015; 75:579-86. [PMID: 25577133 PMCID: PMC4341010 DOI: 10.1007/s00280-014-2671-x] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 12/30/2014] [Indexed: 12/18/2022]
Abstract
Purpose
To define the dose-limiting toxicity (DLT), maximum tolerated dose (MTD) and pharmacokinetics (PK) of PEP02, a novel liposome-encapsulated irinotecan, in patients with advanced refractory solid tumors. Methods Patients were enrolled in cohorts of one to three to receive escalating dose of PEP02 in a phase I trial. PEP02, from 60 to 180 mg/m2, was given as a 90-min intravenous infusion, every 3 weeks. Results A total of 11 patients were enrolled into three dose levels: 60 (one patient), 120 (six patients) and 180 mg/m2 (four patients). DLT was observed in three patients, one at 120 mg/m2 (grade 3 catheter-related infection) and two at 180 mg/m2 (grade 4 neutropenia lasting for >3 days in one, grade 4 hematological toxicities and grade 4 diarrhea in the other). MTD was determined as 120 mg/m2. Comparing with those after free-form irinotecan in the literature, the dose-normalized PK of SN-38 (the active metabolite) after PEP02 was characterized by lower Cmax, prolonged terminal half-life and higher AUC but with significant inter-individual variation. One patient who died of treatment-related toxicity had significantly higher Cmax and AUC levels of SN-38 than those of the other three patients at 180 mg/m2. Post hoc pharmacogenetic study showed that the patient had a combined heterozygosity genotype of UGT1A1*6/*28. Two patients had objective tumor response. Conclusions PEP02 apparently modified the PK parameters of irinotecan and SN-38 by liposome encapsulation. The MTD of PEP02 monotherapy at 3-week interval is 120 mg/m2, which will be the recommended dose for future studies.
Collapse
Affiliation(s)
- T C Chang
- Department of Gynecology, Linkuo Chang-Gung Memorial Hospital, No.5, Fu-Hsing Street, Kuei-shan Hsiang, Taoyuan, 33305, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Zamboni WC, Ramalingam S, Friedland DM, Edwards RP, Stoller RG, Strychor S, Maruca L, Zamboni BA, Belani CP, Ramanathan RK. Phase I and pharmacokinetic study of pegylated liposomal CKD-602 in patients with advanced malignancies. Clin Cancer Res 2009; 15:1466-72. [PMID: 19190127 DOI: 10.1158/1078-0432.ccr-08-1405] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE S-CKD602 is a pegylated liposomal formulation of CKD602, a semisynthetic camptothecin analogue. Pegylated (STEALTH) liposomes can achieve extended drug exposure in plasma and tumor. Based on promising preclinical data, the first phase I study of S-CKD602 was done in patients with refractory solid tumors. EXPERIMENTAL DESIGN S-CKD602 was administered i.v. every 3 weeks. Modified Fibonacci escalation was used (three to six patients/cohort), and dose levels ranged from 0.1 to 2.5 mg/m2. Serial plasma samples were obtained over 2 weeks and total (lactone+hydroxyl acid) concentrations of encapsulated, released, and sum total (encapsulated+released) CKD602 measured by liquid chromatography-tandem mass spectrometry. RESULTS Forty-five patients (21 males) were treated. Median age, 62 years (range, 33-79 years) and Eastern Cooperative Oncology Group status, 0 to 1 (43 patients) and 2 (2 patients). Dose-limiting toxicities of grade 3 mucositis occurred in one of six patients at 0.3 mg/m2, grade 3 and 4 bone marrow suppression in two of three patients at 2.5 mg/m2, and grade 3 febrile neutropenia and anemia in one of six patients at 2.1 mg/m2. The maximum tolerated dose was 2.1 mg/m2. Partial responses occurred in two patients with refractory ovarian cancer (1.7 and 2.1 mg/m2). High interpatient variability occurred in the pharmacokinetic disposition of encapsulated and released CKD602. CONCLUSIONS S-CKD602 represents a promising new liposomal camptothecin analogue with manageable toxicity and promising antitumor activity. Phase II studies of S-CKD602 at 2.1 mg/m2 i.v. once every 3 weeks are planned. Prolonged plasma exposure over 1 to 2 weeks is consistent with STEALTH liposomes and provides extended exposure compared with single doses of nonliposomal camptothecins.
Collapse
Affiliation(s)
- William C Zamboni
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Molecular Therapeutics, Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina 27599-7360, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
|
13
|
Abstract
The biological picture of cancer is rapidly advancing from models built from phenomenological descriptions to network models derived from systems biology, which can capture the evolving pathophysiology of the disease at the molecular level. The translation of this (still academic) picture into a clinically relevant framework can be enabling for the war on cancer, but it is a scientific and technological challenge. In this review, we discuss emerging in vitro diagnostic technologies and therapeutic approaches that are being developed to handle this challenge. Our discussion of in vitro diagnostics is guided by the theme of making large numbers of measurements accurately, sensitively, and at very low cost. We discuss diagnostic approaches based on microfluidics and nanotechnology. We then review the current state of the art of nanoparticle-based therapeutics that have reached the clinic. The goal of the presentation is to identify nanotherapeutic strategies that are designed to increase efficacy while simultaneously minimizing the toxic side effects commonly associated with cancer chemotherapies.
Collapse
Affiliation(s)
- James R Heath
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91125, USA.
| | | |
Collapse
|
14
|
Koizumi F, Kitagawa M, Negishi T, Onda T, Matsumoto SI, Hamaguchi T, Matsumura Y. Novel SN-38-incorporating polymeric micelles, NK012, eradicate vascular endothelial growth factor-secreting bulky tumors. Cancer Res 2006; 66:10048-56. [PMID: 17047068 DOI: 10.1158/0008-5472.can-06-1605] [Citation(s) in RCA: 207] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
7-Ethyl-10-hydroxy-camptothecin (SN-38), a biological active metabolite of irinotecan hydrochloride (CPT-11), has potent antitumor activity but has not been used clinically because it is a water-insoluble drug. For delivery by i.v. injection, we have successfully developed NK012, a SN-38-releasing nanodevice. The purpose of this study is to investigate the pharmacologic character of NK012 as an anticancer agent, especially in a vascular endothelial growth factor (VEGF)-secreting tumor model. The particle size of NK012 was approximately 20 nm with a narrow size distribution. NK012 exhibited a much higher cytotoxic effect against lung and colon cancer cell lines as compared with CPT-11. NK012 showed significantly potent antitumor activity against a human colorectal cancer HT-29 xenograft as compared with CPT-11. Enhanced and prolonged distribution of free SN-38 in the tumor was observed after the injection of NK012. NK012 also had significant antitumor activity against bulky SBC-3/Neo (1,533.1 +/- 1,204.7 mm(3)) and SBC-3/VEGF tumors (1,620.7 +/- 834.0 mm(3)) compared with CPT-11. Furthermore, NK012 eradicated bulky SBC-3/VEGF tumors in all mice but did not eradicate SBC-3/Neo tumors. In the drug distribution analysis, an increased accumulation of SN-38 in SBC-3/VEGF tumors was observed as compared with that in SBC-3/Neo tumors. NK012 markedly enhanced the antitumor activity of SN-38, especially in highly VEGF-secreting tumors, and could be a promising SN-38-based formulation.
Collapse
Affiliation(s)
- Fumiaki Koizumi
- Investigative Treatment Division, Research Center for Innovative Oncology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | | | | | | | | | | | | |
Collapse
|
15
|
Affiliation(s)
- William C Zamboni
- Molecular Therapeutics Drug Discovery Program, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania 15213-1863, USA
| |
Collapse
|