1
|
Geng WC, Jiang ZT, Chen SL, Guo DS. Supramolecular interaction in the action of drug delivery systems. Chem Sci 2024; 15:7811-7823. [PMID: 38817563 PMCID: PMC11134347 DOI: 10.1039/d3sc04585d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 04/27/2024] [Indexed: 06/01/2024] Open
Abstract
Complex diseases and diverse clinical needs necessitate drug delivery systems (DDSs), yet the current performance of DDSs is far from ideal. Supramolecular interactions play a pivotal role in various aspects of drug delivery, encompassing biocompatibility, drug loading, stability, crossing biological barriers, targeting, and controlled release. Nevertheless, despite having some understanding of the role of supramolecular interactions in drug delivery, their incorporation is frequently overlooked in the design and development of DDSs. This perspective provides a brief analysis of the involved supramolecular interactions in the action of drug delivery, with a primary emphasis on the DDSs employed in the clinic, mainly liposomes and polymers, and recognized phenomena in research, such as the protein corona. The supramolecular interactions implicated in various aspects of drug delivery systems, including biocompatibility, drug loading, stability, spatiotemporal distribution, and controlled release, were individually analyzed and discussed. This perspective aims to trigger a comprehensive and systematic consideration of supramolecular interactions in the further development of DDSs. Supramolecular interactions embody the true essence of the interplay between the majority of DDSs and biological systems.
Collapse
Affiliation(s)
- Wen-Chao Geng
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University Tianjin 300071 China
| | - Ze-Tao Jiang
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University Tianjin 300071 China
| | - Shi-Lin Chen
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University Tianjin 300071 China
| | - Dong-Sheng Guo
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University Tianjin 300071 China
| |
Collapse
|
2
|
Wileński S, Koper A, Śledzińska P, Bebyn M, Koper K. Innovative strategies for effective paclitaxel delivery: Recent developments and prospects. J Oncol Pharm Pract 2024; 30:367-384. [PMID: 38204196 DOI: 10.1177/10781552231208978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
PURPOSE Paclitaxel is an effective chemotherapeutic agent against a variety of cancer types. However, the clinical utility of paclitaxel is restricted by its poor solubility in water and high toxicity, resulting in low drug tolerance. These difficulties could be resolved by using suitable pharmacological carriers. Hence, it is essential to determine innovative methods of administering this effective medication to overcome paclitaxel's inherent limitations. METHODS An extensive literature search was conducted using multiple electronic databases to identify relevant studies published. RESULTS In this comprehensive analysis, many different paclitaxel delivery systems are covered and discussed, such as albumin-bound paclitaxel, polymeric micelles, paclitaxel-loaded liposomes, prodrugs, cyclodextrins, and peptide-taxane conjugates. Moreover, the review also covers various delivery routes of conventional paclitaxel or novel paclitaxel formulations, such as oral administration, local applications, and intraperitoneal delivery. CONCLUSION In addition to albumin-bound paclitaxel, polymeric micelles appear to be the most promising formulations for innovative drug delivery systems at present. A variety of variants of polymeric micelles are currently undergoing advanced phases of clinical trials.
Collapse
Affiliation(s)
- Sławomir Wileński
- Department of Pharmaceutical Technology, Nicolaus Copernicus University in Torun, Ludwik Rydygier Collegium Medicum, Bydgoszcz, Poland
- Central Cytostatic Drug Department, Hospital Pharmacy, The F. Lukaszczyk Oncology Centre, Bydgoszcz, Poland
| | - Agnieszka Koper
- Department of Oncology and Brachytherapy, Nicolaus Copernicus University in Torun, Ludwik Rydygier Collegium Medicum, Bydgoszcz, Poland
- Department of Oncology, Franciszek Lukaszczyk Oncology Centre, Bydgoszcz, Poland
| | - Paulina Śledzińska
- Department of Neurosurgery, 10th Military Research Hospital and Polyclinic, Bydgoszcz, Poland
| | - Marek Bebyn
- Department of Neurosurgery, 10th Military Research Hospital and Polyclinic, Bydgoszcz, Poland
| | - Krzysztof Koper
- Department of Oncology, Franciszek Lukaszczyk Oncology Centre, Bydgoszcz, Poland
- Department of Clinical Oncology, and Nursing, Department of Oncological Surgery, Nicolaus Copernicus University in Torun, Ludwik Rydygier Collegium Medicum, Bydgoszcz, Poland
| |
Collapse
|
3
|
Gaughan EM, Horton BJ. Outcomes From Cytotoxic Chemotherapy Following Progression on Immunotherapy in Metastatic Melanoma: An Institutional Case-Series. Front Oncol 2022; 12:855782. [PMID: 35574303 PMCID: PMC9096114 DOI: 10.3389/fonc.2022.855782] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 03/28/2022] [Indexed: 11/29/2022] Open
Abstract
Introduction The role of chemotherapy in the management of advanced melanoma is limited due to low response rates and short survival. Improved outcomes to chemotherapy administered after immunotherapy for metastatic melanoma and other solid tumors have been reported. We studied the outcomes of subjects treated at the University of Virginia (UVA) with chemotherapy following progression on prior systemic immunotherapy and compared the results with the existing literature. Materials and Methods Subjects were identified through an institutional database of patients treated with immunotherapy at UVA. Demographic, pathologic and clinical factors were collected, along with dates of therapy, investigator-assessed best response as per Response Evaluation Criteria for Solid Tumors version 1.1 and dates of death or last follow up. Kaplan-Meier survival estimates and log-rank tests were used to perform time to event analysis of progression free survival and overall survival. Results Forty-five patients were identified who met the inclusion criteria including 24 men and 21 women with a median age of 61 years. All patients had received at least one line of immunotherapy including 64.4% with prior anti-PD1 treatment. The cytotoxic chemotherapy regimens used included carboplatin with paclitaxel (55.6%), temozolomide (31.1%) and nab-paclitaxel (13.3%). The overall response rate for cytotoxic chemotherapy 22.2% and the disease control rate was 35.6%. The median progression-free survival was 1.7 months and median overall survival was 4.7 months. Nineteen (42.2%) patients survived greater than 6 months and seven (15.5%) patients survived over 12 months. Fourteen patients were able to proceed to further therapy. Discussion Our results reveal that receipt of immunotherapy prior to chemotherapy for metastatic melanoma does not appear to improve the benefit of chemotherapy. The palliation of symptoms, maintenance of performance status and disease control may be valuable for some patients during this time of robust research and discovery for metastatic melanoma.
Collapse
Affiliation(s)
- Elizabeth M Gaughan
- Department of Medicine, Division of Hematology and Medical Oncology, University of Virginia, Charlottesville, VA, United States
| | - Bethany J Horton
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
4
|
Moyers JT, Glitza Oliva IC. Immunotherapy for Melanoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1342:81-111. [PMID: 34972963 DOI: 10.1007/978-3-030-79308-1_3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Melanoma is the leading cause of death from skin cancer and is responsible for over 7000 deaths in the USA each year alone. For many decades, limited treatment options were available for patients with metastatic melanoma; however, over the last decade, a new era in treatment dawned for oncologists and their patients. Targeted therapy with BRAF and MEK inhibitors represents an important cornerstone in the treatment of metastatic melanoma; however, this chapter carefully reviews the past and current therapy options available, with a significant focus on immunotherapy-based approaches. In addition, we provide an overview of the results of recent advances in the adjuvant setting for patients with resected stage III and stage IV melanoma, as well as in patients with melanoma brain metastases. Finally, we provide a brief overview of the current research efforts in the field of immuno-oncology for melanoma.
Collapse
Affiliation(s)
- Justin T Moyers
- Department of Investigational Cancer Therapeutics, UT MD Anderson Cancer Center, Houston, TX, USA.,Division of Hematology and Oncology, Department of Medicine, University of California, Irvine, Orange, CA, USA
| | | |
Collapse
|
5
|
Shevtsov M, Kaesler S, Posch C, Multhoff G, Biedermann T. Magnetic nanoparticles in theranostics of malignant melanoma. EJNMMI Res 2021; 11:127. [PMID: 34905138 PMCID: PMC8671576 DOI: 10.1186/s13550-021-00868-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 11/28/2021] [Indexed: 12/14/2022] Open
Abstract
Malignant melanoma is an aggressive tumor with a tendency to metastasize early and with an increasing incidence worldwide. Although in early stage, melanoma is well treatable by excision, the chances of cure and thus the survival rate decrease dramatically after metastatic spread. Conventional treatment options for advanced disease include surgical resection of metastases, chemotherapy, radiation, targeted therapy and immunotherapy. Today, targeted kinase inhibitors and immune checkpoint blockers have for the most part replaced less effective chemotherapies. Magnetic nanoparticles as novel agents for theranostic purposes have great potential in the treatment of metastatic melanoma. In the present review, we provide a brief overview of treatment options for malignant melanoma with different magnetic nanocarriers for theranostics. We also discuss current efforts of designing magnetic particles for combined, multimodal therapies (e.g., chemotherapy, immunotherapy) for malignant melanoma.
Collapse
Affiliation(s)
- Maxim Shevtsov
- Central Institute for Translational Cancer Research (TranslaTUM), Radiation Immuno-Oncology Group, Klinikum rechts der Isar, School of Medicine, Technical University Munich (TUM), Einstein Str. 25, 81675, Munich, Germany
- Laboratory of Biomedical Cell Technologies, Far Eastern Federal University, Primorsky Krai, 690091, Vladivostok, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str, Saint Petersburg, Russian Federation, 197341
| | - Susanne Kaesler
- Department of Dermatology and Allergology, Klinikum rechts der Isar, School of Medicine, Technical University Munich (TUM), Biedersteinerstrasse 29, 80802, Munich, Germany
| | - Christian Posch
- Department of Dermatology and Allergology, Klinikum rechts der Isar, School of Medicine, Technical University Munich (TUM), Biedersteinerstrasse 29, 80802, Munich, Germany
| | - Gabriele Multhoff
- Central Institute for Translational Cancer Research (TranslaTUM), Radiation Immuno-Oncology Group, Klinikum rechts der Isar, School of Medicine, Technical University Munich (TUM), Einstein Str. 25, 81675, Munich, Germany
- Department of Radiation Oncology, Klinikum rechts der Isar, School of Medicine, Technical University Munich (TUM), Ismaninger Str. 22, 81675, Munich, Germany
| | - Tilo Biedermann
- Department of Dermatology and Allergology, Klinikum rechts der Isar, School of Medicine, Technical University Munich (TUM), Biedersteinerstrasse 29, 80802, Munich, Germany.
| |
Collapse
|
6
|
Stromal Protein-Mediated Immune Regulation in Digestive Cancers. Cancers (Basel) 2021; 13:cancers13010146. [PMID: 33466303 PMCID: PMC7795083 DOI: 10.3390/cancers13010146] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/21/2020] [Accepted: 12/24/2020] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Solid cancers are surrounded by a network of non-cancerous cells comprising different cell types, including fibroblasts, and acellular protein structures. This entire network is called the tumor microenvironment (TME) and it provides a physical barrier to the tumor shielding it from infiltrating immune cells, such as lymphocytes, or therapeutic agents. In addition, the TME has been shown to dampen efficient immune responses of infiltrated immune cells, which are key in eliminating cancer cells from the organism. In this review, we will discuss how TME proteins in particular are involved in this dampening effect, known as immunosuppression. We will focus on three different types of digestive cancers: pancreatic cancer, colorectal cancer, and gastric cancer. Moreover, we will discuss current therapeutic approaches using TME proteins as targets to reverse their immunosuppressive effects. Abstract The stromal tumor microenvironment (TME) consists of immune cells, vascular and neural structures, cancer-associated fibroblasts (CAFs), as well as extracellular matrix (ECM), and favors immune escape mechanisms promoting the initiation and progression of digestive cancers. Numerous ECM proteins released by stromal and tumor cells are crucial in providing physical rigidity to the TME, though they are also key regulators of the immune response against cancer cells by interacting directly with immune cells or engaging with immune regulatory molecules. Here, we discuss current knowledge of stromal proteins in digestive cancers including pancreatic cancer, colorectal cancer, and gastric cancer, focusing on their functions in inhibiting tumor immunity and enabling drug resistance. Moreover, we will discuss the implication of stromal proteins as therapeutic targets to unleash efficient immunotherapy-based treatments.
Collapse
|
7
|
Wang X, Xu W, Chi Z, Si L, Sheng X, Kong Y, Zhou L, Mao L, Lian B, Tang B, Yan X, Bai X, Cui C, Guo J. Chemotherapy combined with antiangiogenic drugs as salvage therapy in advanced melanoma patients progressing on PD-1 immunotherapy. Transl Oncol 2020; 14:100949. [PMID: 33221685 PMCID: PMC7689327 DOI: 10.1016/j.tranon.2020.100949] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 10/31/2020] [Accepted: 11/05/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND This study aimed to evaluate the effect of salvage therapy with nab-paclitaxel (nab-p) or temozolomide (TMZ) combined with antiangiogenic drugs in programmed death 1 (PD-1) inhibitor-resistant patients with unresectable metastatic melanoma. METHODS We conducted a retrospective review of 69 metastatic melanoma patients who received nab-p or TMZ combined with antiangiogenic drugs after developing PD-1 inhibitor resistance and were treated at the Beijing Cancer Hospital between 2016 and 2019. The disease control rate (c-DCR) and progression-free survival (c-PFS) of salvage CA (chemotherapy combined with antiangiogenic drugs) regimens were investigated. Univariate and multivariate analyses were performed to evaluate the clinical pathological factors affecting the outcomes. Then, a nomogram was formulated to predict the probability of 3-month and 6-month c-PFS based on the multivariate analysis results. RESULTS The c-DCR was 63.8%, and the median c-PFS was 3.0 months. In the univariate analysis, factors associated with the c-DCR were included the melanoma subtype, baseline platelet-to-lymphocyte ratio (PLR) and best response status to PD-1 inhibitors. Factors influencing c-PFS included age, baseline lactic dehydrogenase, PLR, neutrophil-to-lymphocyte ratio (NLR), PFS duration of anti-PD-1 therapy (p-PFS), and the best response and progression pattern of PD-1 inhibitors. In the multivariate analysis, age <65 years, heterogeneous progression pattern and baseline PLR<200 were significantly associated with improved c-PFS. The concordance index (C-index) of the nomogram was equal to 0.65 (95% CI 0.566-0.734). CONCLUSIONS CA regimens demonstrated promising effects in PD-1 inhibitor-resistant patients. The nomogram could be a valuable predictive module for salvage therapy choice in PD-1 inhibitor-resistant patients.
Collapse
Affiliation(s)
- Xuan Wang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Weiran Xu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Zhihong Chi
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Lu Si
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Xinan Sheng
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Yan Kong
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Li Zhou
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Lili Mao
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Bin Lian
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Bixia Tang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Xieqiao Yan
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Xue Bai
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Chuanliang Cui
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Jun Guo
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China.
| |
Collapse
|
8
|
Tang Y, Chen M, Xie Q, Li L, Zhu L, Ma Q, Gao S. Construction and evaluation of hyaluronic acid-based copolymers as a targeted chemotherapy drug carrier for cancer therapy. NANOTECHNOLOGY 2020; 31:305702. [PMID: 32272454 DOI: 10.1088/1361-6528/ab884d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Melanoma (MM) is a highly aggressive skin cancer with limited treatment options. Although chemotherapy has been using for advanced melanoma treatment, the lack of targetability, the poor biocompatibility and the severe side effects still hamper the wide applications of chemotherapy agents in MM management. Herein, a biocompatible and biodegradable polymeric hyaluronic acid nanoparticle (HANP) encapsulated with Paclitaxel (PTX) was developed for MM targeted therapy. Our results showed that PTX at 37 ± 2.1% (w/w) was able to be loaded into HANP with over 5 d of stability under physiological conditions. In vitro, HANP/PTX presented hyaluronidase-dependent drug release. Compared to free PTX, HANP/PTX demonstrated a 6-75 times higher growth inhibition in five different cancer cells, while only presenting minimum toxicity to normal cells. After intravenous administration at a 10 mg kg-1 equivalent dose of PTX, HANP/PTX significantly ablated MM tumor growth in a mouse model. As confirmed by 18F-fluoro-2-deoxy-D-glucose (FDG) positron emission tomography (PET) imaging, the tumors started to respond to the HANP/PTX as early as 7 d after the initial treatment, which will significantly benefit for personalized treatment. In conclusion, the HANP/PTX nanocomplex demonstrated great promise as a translational nanomedicine for cancer chemotherapy.
Collapse
Affiliation(s)
- Yuting Tang
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun 130033, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
9
|
Beiu C, Giurcaneanu C, Grumezescu AM, Holban AM, Popa LG, Mihai MM. Nanosystems for Improved Targeted Therapies in Melanoma. J Clin Med 2020; 9:jcm9020318. [PMID: 31979325 PMCID: PMC7073828 DOI: 10.3390/jcm9020318] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/11/2020] [Accepted: 01/20/2020] [Indexed: 02/06/2023] Open
Abstract
Melanoma is one of the most aggressive forms of skin cancer, with limited therapeutic options. Since its incidence has been rapidly rising in recent years, the study of new targeted therapeutic strategies has increased. The implication of nanoscience in the development of alternative targeted therapies for melanoma has multiple benefits and could significantly improve the outcome of melanoma patients. In this paper, we review the most recent progress in the field of targeted therapies, emphasizing the impact of nanoscale materials on the targeting and controlled release of anti-tumor drugs. The applications of nanomedicine in the management of melanoma are extensive and refer to sentinel lymph node mapping, chemotherapy, and RNA interference; each of these applications harboring the potential to develop efficient and personalized diagnostic techniques and therapies. Further research, especially in clinical trials, is needed to establish whether fighting melanoma on the nanoscale level represents the key to reaching a critical inflection point in mankind’s battle with metastatic melanoma.
Collapse
Affiliation(s)
- Cristina Beiu
- Department of Oncologic Dermatology-“Elias” Emergency University Hospital, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.B.); (C.G.); (L.G.P.)
| | - Calin Giurcaneanu
- Department of Oncologic Dermatology-“Elias” Emergency University Hospital, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.B.); (C.G.); (L.G.P.)
| | - Alexandru Mihai Grumezescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 1–7 Polizu Street, 011061 Bucharest, Romania;
| | - Alina Maria Holban
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 1–7 Polizu Street, 011061 Bucharest, Romania;
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, 030018 Bucharest, Romania
- Correspondence: ; Tel.: +40-721-600-737
| | - Liliana Gabriela Popa
- Department of Oncologic Dermatology-“Elias” Emergency University Hospital, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.B.); (C.G.); (L.G.P.)
| | - Mara Mădălina Mihai
- Department of Oncologic Dermatology-“Elias” Emergency University Hospital, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.B.); (C.G.); (L.G.P.)
| |
Collapse
|
10
|
Abstract
Nanomedicines have historically struggled to find clinical relevance and to achieve translation successfully. In this Perspective, we discuss possible reasons for this difficulty and highlight several key features of nanomedicines that are often overlooked by biomedical engineers. We present the notion of clinical multifunctionality as distinct from multifunctionality as it is traditionally described at the nanoscale and emphasize its importance through examples of nanomedicines that have demonstrated emergent clinical multifunctionality once translated. We also describe a phenomenon in which clinical multifunctionality results in diagonal translation after a nanomedicine is adopted by clinicians to serve as a solution for a clinical problem that it was not designed to solve. Biomedical engineers can take advantage of these phenomena to assist in achieving clinical translation of new nanomedicines.
Collapse
Affiliation(s)
- Keegan Guidolin
- Department of Surgery , University of Toronto , Toronto M5G 1L7 , Canada
- Institute of Biomaterials and Biomedical Engineering , University of Toronto , Toronto M5G 1L7 , Canada
- Princess Margaret Cancer Centre , Toronto M5G 1L7 , Canada
| | - Gang Zheng
- Institute of Biomaterials and Biomedical Engineering , University of Toronto , Toronto M5G 1L7 , Canada
- Department of Medical Biophysics , University of Toronto , Toronto M5G 1L7 , Canada
- Princess Margaret Cancer Centre , Toronto M5G 1L7 , Canada
| |
Collapse
|
11
|
Madamsetty VS, Paul MK, Mukherjee A, Mukherjee S. Functionalization of Nanomaterials and Their Application in Melanoma Cancer Theranostics. ACS Biomater Sci Eng 2019; 6:167-181. [PMID: 33463233 DOI: 10.1021/acsbiomaterials.9b01426] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Treatment and cure for melanoma, the most aggressive subcategory of skin cancer, still remains a daunting challenge to be circumvented. When metastasized, it requires radiotherapy, chemotherapy, targeted therapy, immunotherapy, etc. as its treatment, although it can be removed by surgical intervention if detected in its early stage. Development of upgraded therapeutic modalities for melanoma facilitating early diagnosis with subsequent excision before metastasis is, therefore, an urgent need. As we witnessed, nanotechnology has become instrumental with its far-reaching ramifications both in diagnosis and treatment of melanoma. In this review we are going to summarize the encouraging developments made in recent times for functionalization of nanoparticles (including liposomes, polymeric, metal, viral, protein nanoparticles) to create numerous theranostics (therapy plus diagnostics) for melanoma. We will also reflect on the melanoma statistics, molecular biology, conventional therapies, ongoing clinical trials, and future outlook.
Collapse
Affiliation(s)
- Vijay Sagar Madamsetty
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville 32224, Florida, United States
| | - Manash K Paul
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, The University of California, Los Angeles, Factor Building 621 Charles E. Young Drive, Los Angeles 90095, California, United States
| | - Anubhab Mukherjee
- Sealink Pharmaceuticals, Trendz Avenue, First floor, Plot Number 12, Gafoor Nagar, Madhapur, Hyderabad 500081, India
| | - Sudip Mukherjee
- Department of Bioengineering, Rice University, Houston 77030, Texas, United States
| |
Collapse
|
12
|
Ko G, Kim T, Ko E, Park D, Lee Y. Synergistic Enhancement of Paclitaxel-induced Inhibition of Cell Growth by Metformin in Melanoma Cells. Dev Reprod 2019; 23:119-128. [PMID: 31321352 PMCID: PMC6635613 DOI: 10.12717/dr.2019.23.2.119] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 05/27/2019] [Accepted: 06/08/2019] [Indexed: 11/25/2022]
Abstract
Melanoma is one of the most aggressive and treatment-resistant malignancies.
Antidiabetic drug metformin has been reported to inhibit cell proliferation and
metastasis in many cancers, including melanoma. Metformin suppresses the
mammalian target of rapamycin (mTOR) and our previous study showed that it also
inhibits the activity of extracellular signal-regulated kinase (ERK). Paclitaxel
is currently prescribed for treatment of melanoma. However, paclitaxel induced
the activation of ERK/mitogen-activated protein kinase (MAPK) pathway, a cell
signaling pathway implicated in cell survival and proliferation. Therefore, we
reasoned that combined treatment of paclitaxel with metformin could be more
effective in the suppression of cell proliferation than treatment of paclitaxel
alone. Here, we investigated the combinatory effect of paclitaxel and metformin
on the cell survival in SK-MEL-28 melanoma cell line. Our study shows that the
combination of paclitaxel and metformin has synergistic effect on cell survival
and suppresses the expression of proteins involved in cancer metastasis. These
findings suggest that the combination of paclitaxel and metformin can be a
possible therapeutic option for treatment of melanoma.
Collapse
Affiliation(s)
- Gihyun Ko
- Dept. of Medicine, Jeju National University School of Medicine, Jeju 63243, Korea
| | - Taehyung Kim
- Dept. of Medicine, Jeju National University School of Medicine, Jeju 63243, Korea
| | - Eunjeong Ko
- Dept. of Medicine, Jeju National University School of Medicine, Jeju 63243, Korea
| | - Deokbae Park
- Histology, Jeju National University School of Medicine, Jeju 63243, Korea
| | - Youngki Lee
- Histology, Jeju National University School of Medicine, Jeju 63243, Korea
| |
Collapse
|
13
|
Fan Y, Mao Y, Cao S, Xia G, Zhang Q, Zhang H, Qiu F, Kang N. S5, a Withanolide Isolated from Physalis Pubescens L., Induces G2/M Cell Cycle Arrest via the EGFR/P38 Pathway in Human Melanoma A375 Cells. Molecules 2018; 23:E3175. [PMID: 30513793 PMCID: PMC6321527 DOI: 10.3390/molecules23123175] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 11/25/2018] [Accepted: 11/29/2018] [Indexed: 11/16/2022] Open
Abstract
S5 is a withanolide natural product isolated from Physalis pubescens L. Our previous experimental studies found that it has significant antitumor activity on renal cell carcinoma. In the present study, the anti-melanoma effect of S5 and the related molecular mechanism was first investigated. It was found that S5 induced an obvious growth inhibitory effect on human melanoma A375 cells with low toxicity to human peripheral blood cells. Furthermore, the results demonstrated that the cell death mode of S5 on A375 cells is not due to inducing apoptosis and autophagy. However, there was a significant time-dependent increase in G2/M phase after treatment of A375 with S5. Meanwhile, S5 could also decrease the protein expression of Cdc25c, Cdc2, and CyclinB1, and increased the expression of p-P53 and P21, suggesting that S5 inhibited A375 cell death through G2/M phase arrest. Moreover, the signal pathway factors P38, extracellular regulated protein kinases (ERK), and epidermal growth factor receptor (EGFR) were observed taking part in the S5-induced A375 cells growth inhibitory effect. In addition, suppressing P38 and EGFR reversed the cell proliferation inhibitory effect and G2/M cell cycle arrest induced by S5 and inhibition of EGFR enhanced the downregulation of the expression of P38 and p-P38, indicating that S5 induced A375 G2/M arrest through the EGFR/P38 pathway. Briefly, this study explained for the first time the mechanism of S5-induced A375 cell growth inhibition in order to provide the basis for its clinical application in melanoma.
Collapse
Affiliation(s)
- Yuqi Fan
- Department of Biochemistry, School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| | - Yiwei Mao
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| | - Shijie Cao
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| | - Guiyang Xia
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
- Department of Pharmaceutical Chemistry, School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| | - Qiang Zhang
- Department of Biochemistry, School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| | - Hongyang Zhang
- Department of Biochemistry, School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| | - Feng Qiu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
- Department of Pharmaceutical Chemistry, School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| | - Ning Kang
- Department of Biochemistry, School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| |
Collapse
|
14
|
A Contemporary Review of the Treatment Landscape and the Role of Predictive and Prognostic Biomarkers in Pancreatic Adenocarcinoma. Can J Gastroenterol Hepatol 2018; 2018:1863535. [PMID: 29623263 PMCID: PMC5829312 DOI: 10.1155/2018/1863535] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 01/17/2018] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer continues to represent one of the leading causes of cancer-related morbidity and mortality in the developed world. Over the past decade, novel systemic therapy combination regimens have contributed to clinically meaningful and statistically significant improvements in overall survival as compared to conventional monotherapy. However, the prognosis for most patients remains guarded secondary to the advanced stages of disease at presentation. There is growing consensus that outcomes can be further optimized with the use of predictive and prognostic biomarkers whereby the former can be enriching for patients who would benefit from therapies and the latter can inform decision-making regarding the need and timing of advanced care planning. One of the challenges of current biomarkers is the lack of standardization across clinical practices such that comparability between jurisdictions can be difficult or even impossible. This inconsistency can impede widespread implementation of their use. In this review article, we provide a comprehensive overview of the contemporary treatment options for pancreatic cancer and we offer some insights into the existing landscape and future directions of biomarker development for this disease.
Collapse
|
15
|
Integrin β1 activation induces an anti-melanoma host response. PLoS One 2017; 12:e0175300. [PMID: 28448494 PMCID: PMC5407755 DOI: 10.1371/journal.pone.0175300] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 03/23/2017] [Indexed: 11/29/2022] Open
Abstract
TGF-β is a cytokine thought to function as a tumor promoter in advanced malignancies. In this setting, TGF-β increases cancer cell proliferation, survival, and migration, and orchestrates complex, pro-tumorigenic changes in the tumor microenvironment. Here, we find that in melanoma, integrin β1-mediated TGF-β activation may also produce tumor suppression via an altered host response. In the A375 human melanoma cell nu/nu xenograft model, we demonstrate that cell surface integrin β1-activation increases TGF-β activity, resulting in stromal activation, neo-angiogenesis and, unexpectedly for this nude mouse model, increase in the number of intra-tumoral CD8+ T lymphocytes within the tumor microenvironment. This is associated with attenuation of tumor growth and long-term survival benefit. Correspondingly, in human melanomas, TGF-β1 correlates with integrin β1/TGF-β1 activation and the expression of markers for vasculature and stromal activation. Surprisingly, this integrin β1/TGF-β1 transcriptional footprint also correlates with the expression of markers for tumor-infiltrating lymphocytes, multiple immune checkpoints and regulatory pathways, and, importantly, better long-term survival of patients. These correlations are unique to melanoma, in that we do not observe similar associations between β1 integrin/TGF-β1 activation and better long-term survival in other human tumor types. These results suggest that activation of TGF-β1 in melanoma may be associated with the generation of an anti-tumor host response that warrants further study.
Collapse
|
16
|
Nevala WK, Butterfield JT, Sutor SL, Knauer DJ, Markovic SN. Antibody-targeted paclitaxel loaded nanoparticles for the treatment of CD20 + B-cell lymphoma. Sci Rep 2017; 7:45682. [PMID: 28378801 PMCID: PMC5381215 DOI: 10.1038/srep45682] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 03/01/2017] [Indexed: 12/23/2022] Open
Abstract
We developed a nano-antibody targeted chemotherapy (nATC) delivery strategy in which tumor specific and clinically relevant antibodies (rituximab, anti-CD20) are non-covalently bound to the albumin scaffold of nab-paclitaxel (ABX). We define the nanoparticle formed when the 2 drugs are bound (AR160). The newly created nATC retains the cytotoxicity of ABX and CD20 affinity of rituximab in vitro. We describe the binding characteristics of the ABX and rituximab in AR160 using peptide mapping/Biacore approach. Flow-based methods, including ImageStream and nanoparticle tracking, were used to characterize the AR160 particles in vitro. A mouse model of human B-cell lymphoma was utilized to test in vivo efficacy of AR160 therapy, which suggested improved tumor targeting (biodistribution) as the most likely mechanism of AR160 therapeutic superiority over ABX or rituximab alone. These data suggest a novel platform for nATC delivery using a slight modification of existing cancer drugs with significantly improved treatment efficacy.
Collapse
Affiliation(s)
- Wendy K Nevala
- Department of Oncology Research, Mayo Clinic, 200 1st St SW Rochester, MN 55905, USA
| | - John T Butterfield
- Department of Oncology Research, Mayo Clinic, 200 1st St SW Rochester, MN 55905, USA
| | - Shari L Sutor
- Department of Oncology Research, Mayo Clinic, 200 1st St SW Rochester, MN 55905, USA
| | - Daniel J Knauer
- Department of Oncology Research, Mayo Clinic, 200 1st St SW Rochester, MN 55905, USA
| | - Svetomir N Markovic
- Department of Oncology Research, Mayo Clinic, 200 1st St SW Rochester, MN 55905, USA
| |
Collapse
|
17
|
Drewes CC, Fiel LA, Bexiga CG, Asbahr ACC, Uchiyama MK, Cogliati B, Araki K, Guterres SS, Pohlmann AR, Farsky SP. Novel therapeutic mechanisms determine the effectiveness of lipid-core nanocapsules on melanoma models. Int J Nanomedicine 2016; 11:1261-79. [PMID: 27099491 PMCID: PMC4821388 DOI: 10.2147/ijn.s101543] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Melanoma is a severe metastatic skin cancer with poor prognosis and no effective treatment. Therefore, novel therapeutic approaches using nanotechnology have been proposed to improve therapeutic effectiveness. Lipid-core nanocapsules (LNCs), prepared with poly(ε-caprolactone), capric/caprylic triglyceride, and sorbitan monostearate and stabilized by polysorbate 80, are efficient as drug delivery systems. Here, we investigated the effects of acetyleugenol-loaded LNC (AcE-LNC) on human SK-Mel-28 melanoma cells and its therapeutic efficacies on melanoma induced by B16F10 in C57B6 mice. LNC and AcE-LNC had z-average diameters and zeta potential close to 210 nm and -10.0 mV, respectively. CytoViva(®) microscopy images showed that LNC and AcE-LNC penetrated into SK-Mel-28 cells, and remained in the cytoplasm. AcE-LNC in vitro treatment (18-90×10(9) particles/mL; 1 hour) induced late apoptosis and necrosis; LNC and AcE-LNC (3-18×10(9) particles/mL; 48 hours) treatments reduced cell proliferation and delayed the cell cycle. Elevated levels of nitric oxide were found in supernatant of LNC and AcE-LNC, which were not dependent on nitric oxide synthase expressions. Daily intraperitoneal or oral treatment (days 3-10 after tumor injection) with LNC or AcE-LNC (1×10(12) particles/day), but not with AcE (50 mg/kg/day, same dose as AcE-LNC), reduced the volume of the tumor; nevertheless, intraperitoneal treatment caused toxicity. Oral LNC treatment was more efficient than AcE-LNC treatment. Moreover, oral treatment with nonencapsulated capric/caprylic triglyceride did not inhibit tumor development, implying that nanocapsule supramolecular structure is important to the therapeutic effects. Together, data herein presented highlight the relevance of the supramolecular structure of LNCs to toxicity on SK-Mel-28 cells and to the therapeutic efficacy on melanoma development in mice, conferring novel therapeutic mechanisms to LNC further than a drug delivery system.
Collapse
Affiliation(s)
- Carine C Drewes
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Luana A Fiel
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Celina G Bexiga
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ana Carolina C Asbahr
- Postgraduate Program in Pharmaceutical Nanotechnology, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Mayara K Uchiyama
- Department of Fundamental Chemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Bruno Cogliati
- Department of Pathology, Faculty of Veterinary Medicine, University of São Paulo, São Paulo, Brazil
| | - Koiti Araki
- Department of Fundamental Chemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Sílvia S Guterres
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
- Postgraduate Program in Pharmaceutical Nanotechnology, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Adriana R Pohlmann
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
- Postgraduate Program in Pharmaceutical Nanotechnology, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
- Department of Organic Chemistry, Institute of Chemistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Sandra P Farsky
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
18
|
Hersh EM, Del Vecchio M, Brown MP, Kefford R, Loquai C, Testori A, Bhatia S, Gutzmer R, Conry R, Haydon A, Robert C, Ernst S, Homsi J, Grob JJ, Kendra K, Agarwala SS, Li M, Clawson A, Brachmann C, Karnoub M, Elias I, Renschler MF, Hauschild A. A randomized, controlled phase III trial of nab-Paclitaxel versus dacarbazine in chemotherapy-naïve patients with metastatic melanoma. Ann Oncol 2015; 26:2267-74. [PMID: 26410620 PMCID: PMC6279094 DOI: 10.1093/annonc/mdv324] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 05/08/2015] [Accepted: 07/21/2015] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The efficacy and safety of nab-paclitaxel versus dacarbazine in patients with metastatic melanoma was evaluated in a phase III randomized, controlled trial. PATIENTS AND METHODS Chemotherapy-naïve patients with stage IV melanoma received nab-paclitaxel 150 mg/m(2) on days 1, 8, and 15 every 4 weeks or dacarbazine 1000 mg/m(2) every 3 weeks. The primary end point was progression-free survival (PFS) by independent radiologic review; the secondary end point was overall survival (OS). RESULTS A total of 529 patients were randomized to nab-paclitaxel (n = 264) or dacarbazine (n = 265). Baseline characteristics were well balanced. The majority of patients were men (66%), had an Eastern Cooperative Oncology Group status of 0 (71%), and had M1c stage disease (65%). The median PFS (primary end point) was 4.8 months with nab-paclitaxel and 2.5 months with dacarbazine [hazard ratio (HR), 0.792; 95.1% confidence interval (CI) 0.631-0.992; P = 0.044]. The median OS was 12.6 months with nab-paclitaxel and 10.5 months with dacarbazine (HR, 0.897; 95.1% CI 0.738-1.089; P = 0.271). Independently assessed overall response rate was 15% versus 11% (P = 0.239), and disease control rate (DCR) was 39% versus 27% (P = 0.004) for nab-paclitaxel versus dacarbazine, respectively. The most common grade ≥3 treatment-related adverse events were neuropathy (nab-paclitaxel, 25% versus dacarbazine, 0%; P < 0.001), and neutropenia (nab-paclitaxel, 20% versus dacarbazine, 10%; P = 0.004). There was no correlation between secreted protein acidic and rich in cysteine (SPARC) status and PFS in either treatment arm. CONCLUSIONS nab-Paclitaxel significantly improved PFS and DCR compared with dacarbazine, with a manageable safety profile.
Collapse
Affiliation(s)
- E M Hersh
- Department of Medicine, Arizona Cancer Center, Tucson, USA
| | - M Del Vecchio
- Department of Medical Oncology, Fondazione IRCCS National Tumor Institute, Milan, Italy
| | - M P Brown
- Cancer Clinical Trials Unit, Royal Adelaide Hospital and School of Medicine, University of Adelaide, Adelaide
| | - R Kefford
- Sydney West Cancer Trials Centre/Westmead Hospital and Melanoma Institute Australia, University of Sydney, North Sydney, Australia
| | - C Loquai
- Department of Dermatology, University of Mainz, Mainz, Germany
| | - A Testori
- Melanoma and Muscle Cutaneous Sarcoma Division, European Institute of Oncology, Milan, Italy
| | - S Bhatia
- Department of Medicine, Seattle Cancer Care Alliance, Seattle, USA
| | - R Gutzmer
- Department of Dermatology and Oncology, Hannover Medical School, Hannover, Germany
| | - R Conry
- Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, USA
| | - A Haydon
- Department of Medical Oncology, Alfred Hospital, Melbourne, Australia
| | - C Robert
- Demartology Unit, Department of Medicine, The Gustave Roussy Cancer Institute, Villejuif, France
| | - S Ernst
- Department of Medical Oncology, London Health Sciences Center-London Regional Cancer Program, London, Canada
| | - J Homsi
- Department of Medical Oncology, Banner MD Anderson Cancer Center, Gilbert, USA
| | - J J Grob
- Department of Dermatology, Timone Hospital, APHM and Aix-Marseille University, Marseille, France
| | - K Kendra
- Department of Internal Medicine, Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus
| | - S S Agarwala
- Department of Hematology and Oncology, St Luke's Cancer Center and Temple University, Bethlehem
| | - M Li
- Biometrics and Data Operations/Translational Medicine/Biometrics and Data Operations/Clinical Research & Development/Global Medical Affairs, Celgene Corporation, Summit, USA
| | - A Clawson
- Biometrics and Data Operations/Translational Medicine/Biometrics and Data Operations/Clinical Research & Development/Global Medical Affairs, Celgene Corporation, Summit, USA
| | - C Brachmann
- Biometrics and Data Operations/Translational Medicine/Biometrics and Data Operations/Clinical Research & Development/Global Medical Affairs, Celgene Corporation, Summit, USA
| | - M Karnoub
- Biometrics and Data Operations/Translational Medicine/Biometrics and Data Operations/Clinical Research & Development/Global Medical Affairs, Celgene Corporation, Summit, USA
| | - I Elias
- Biometrics and Data Operations/Translational Medicine/Biometrics and Data Operations/Clinical Research & Development/Global Medical Affairs, Celgene Corporation, Summit, USA
| | - M F Renschler
- Biometrics and Data Operations/Translational Medicine/Biometrics and Data Operations/Clinical Research & Development/Global Medical Affairs, Celgene Corporation, Summit, USA
| | - A Hauschild
- Department of Dermatology, University Medical Center Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
19
|
Kwek SS, Kahn J, Greaney SK, Lewis J, Cha E, Zhang L, Weber RW, Leonard L, Markovic SN, Fong L, Spitler LE. GM-CSF and ipilimumab therapy in metastatic melanoma: Clinical outcomes and immunologic responses. Oncoimmunology 2015; 5:e1101204. [PMID: 27141383 DOI: 10.1080/2162402x.2015.1101204] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 09/23/2015] [Accepted: 09/23/2015] [Indexed: 12/22/2022] Open
Abstract
We conducted a phase II clinical trial of anti-CTLA-4 antibody (ipilimumab) and granulocyte-macrophage colony-stimulating factor (GM-CSF) in 22 patients with metastatic melanoma and determined clinical outcomes and immunologic responses. The treatment consisted of a 3-mo induction with ipilimumab at 10 mg/kg administered every 3 weeks for four doses in combination with GM-CSF at 125 µg/m2 for 14 d beginning on the day of the ipilimumab infusion and then GM-CSF for 3 mo on the same schedule without ipilimumab. This was followed by maintenance therapy with the combination every 3 mo for up to 2 y or until disease progression or unacceptable toxicity. Blood samples for determination of immune subsets were obtained before treatment, at week 3 (end of cycle 1) and at week 6 (end of cycle 2). Blood samples were also obtained from seven subjects who were cancer-free. The immune response disease control (irDC) rate at 24 weeks was 41% and the overall response rate (ORR) was 32%. The median progression free-survival (PFS) was 3.5 mo and the median overall survival (OS) was 21.1 mo. 41% of the patients experienced Grade 3 to 4 adverse events. We conclude that this combination is safe and the results suggest the combination may be more effective than ipilimumab monotherapy. Further, the results suggest that lower levels of CD4+ effector T cells but higher levels of CD8+ T cells expressing PD-1 at pre-treatment could be a potential biomarker for disease control in patients who receive immunotherapy with ipilimumab and GM-CSF. Further trials of this combination are warranted.
Collapse
Affiliation(s)
- Serena S Kwek
- Department of Medicine, Hem/Onc Division, University of California San Francisco , San Francisco, CA, USA
| | - James Kahn
- Northern California Melanoma Center, Saint Mary's Medical Center , San Francisco, CA, USA
| | - Samantha K Greaney
- Northern California Melanoma Center, Saint Mary's Medical Center , San Francisco, CA, USA
| | - Jera Lewis
- Department of Medicine, Hem/Onc Division, University of California San Francisco , San Francisco, CA, USA
| | - Edward Cha
- Department of Medicine, Hem/Onc Division, University of California San Francisco , San Francisco, CA, USA
| | - Li Zhang
- Department of Medicine, Hem/Onc Division, University of California San Francisco , San Francisco, CA, USA
| | - Robert W Weber
- Northern California Melanoma Center, Saint Mary's Medical Center , San Francisco, CA, USA
| | - Lonnie Leonard
- Northern California Melanoma Center, Saint Mary's Medical Center , San Francisco, CA, USA
| | - Svetomir N Markovic
- Division of Hematology, Oncology Department , Mayo Clinic , Rochester, MN, USA
| | - Lawrence Fong
- Department of Medicine, Hem/Onc Division, University of California San Francisco , San Francisco, CA, USA
| | - Lynn E Spitler
- Northern California Melanoma Center, Saint Mary's Medical Center , San Francisco, CA, USA
| |
Collapse
|
20
|
Cid-Arregui A, Juarez V. Perspectives in the treatment of pancreatic adenocarcinoma. World J Gastroenterol 2015; 21:9297-9316. [PMID: 26309356 PMCID: PMC4541382 DOI: 10.3748/wjg.v21.i31.9297] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 05/12/2015] [Accepted: 07/18/2015] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an incurable lethal disease whose incidence rate is growing. There is no effective screening for detection of early stage tumors and, in most cases, PDAC is diagnosed at advanced disease stages, when radical pancreatic resection is not possible. The aggressive nature of pancreatic tumor cells lies in the complex genetic mechanisms behind their uncontrolled capability to grow and metastasize, which involve essential adaptive changes in cellular metabolism, signaling, adhesion and immunoediting. In addition, PDAC cells promote a dense functional stroma that facilitates tumor resistance to chemotherapy and radiation. During the last two decades, gemcitabine has been the reference for the systemic treatment of PDAC. However, recently, a regimen combining fluorouracil, irinotecan, oxaliplatin, and leucovorin (FOLFIRINOX) and another combining albumin-bound paclitaxel with gemcitabine have shown clear therapeutic advantage in advanced PDAC, with survival outcomes of 11.3 and 8.5 mo on phase III trials, respectively, over single-agent gemcitabine. With the pending issue of their higher toxicities, these regimens set the reference for ongoing and future clinical studies in advanced PDAC. In addition, the efficacy of oral fluoropyrimidine (S-1) has been well documented in Asiatic PDAC patients. The development of therapeutic approaches other than cytotoxic drugs has proven difficult in the past, with only one drug (erlotinib) approved to date. Besides, a number of agents targeting signaling pathways in tumor or stroma cells are being investigated. Likewise, immunotherapies that target PDAC in various ways are the subject of a number of clinical trials. The search for reliable biomarkers with diagnostic and prognostic value using genomics and mass spectrometry methods may facilitate monitoring and refinement of therapies. This review focuses on current understanding of the pathogenesis of PDAC and the latest developments in the treatment of advanced PDAC.
Collapse
|
21
|
Kundranda MN, Niu J. Albumin-bound paclitaxel in solid tumors: clinical development and future directions. Drug Des Devel Ther 2015; 9:3767-77. [PMID: 26244011 PMCID: PMC4521678 DOI: 10.2147/dddt.s88023] [Citation(s) in RCA: 169] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Albumin-bound paclitaxel (nab-paclitaxel) is a solvent-free formulation of paclitaxel that was initially developed more than a decade ago to overcome toxicities associated with the solvents used in the formulation of standard paclitaxel and to potentially improve efficacy. Nab-paclitaxel has demonstrated an advantage over solvent-based paclitaxel by being able to deliver a higher dose of paclitaxel to tumors and decrease the incidence of serious toxicities, including severe allergic reactions. To date, nab-paclitaxel has been indicated for the treatment of three solid tumors in the USA. It was first approved for the treatment of metastatic breast cancer in 2005, followed by locally advanced or metastatic non-small-cell lung cancer in 2012, and most recently for metastatic pancreatic cancer in 2013. Nab-paclitaxel is also under investigation for the treatment of a number of other solid tumors. This review highlights key clinical efficacy and safety outcomes of nab-paclitaxel in the solid tumors for which it is currently indicated, discusses ongoing trials that may provide new data for the expansion of nab-paclitaxel's indications into other solid tumors, and provides a clinical perspective on the use of nab-paclitaxel in practice.
Collapse
Affiliation(s)
| | - Jiaxin Niu
- Department of Medical Oncology, Cancer Treatment Centers of America, Goodyear, AZ, USA
| |
Collapse
|
22
|
Mundra V, Peng Y, Kumar V, Li W, Miller DD, Mahato RI. Systemic delivery of nanoparticle formulation of novel tubulin inhibitor for treating metastatic melanoma. Drug Deliv Transl Res 2015; 5:199-208. [PMID: 25924699 DOI: 10.1007/s13346-015-0226-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Clinical translation of tubulin inhibitors for treating melanoma is limited by multidrug efflux transporters, poor aqueous solubility, and dose-limiting peripheral toxicities. Tubulin inhibitors with efficacy in taxane-resistant cancers are promising drug candidates and can be used as single agent or in conjunction with other chemotherapy. Systemic therapy of such a novel tubulin inhibitor, 2-(1H-indol-5-yl)thiazol-4-yl)3,4,5-trimethoxyphenyl methanone (abbreviated as LY293), is limited by its poor aqueous solubility. The objective of this study was to design a polymeric nanocarrier for systemic administration of LY293 to improve tumor accumulation and reduce side effects of tubulin inhibitor in a lung metastasis melanoma mouse model. Methoxy polyethylene glycol-b-poly(carbonate-co-lactide) (mPEG-b-P(CB-co-LA)) random copolymer was synthesized and characterized by ¹H NMR and gel permeation chromatography (GPC). Polymeric nanoparticles were formulated using oil/water (o/w) emulsification method with a mean particle size of 150 nm and loading efficiency of 7.40%. Treatment with LY293-loaded nanoparticles effectively inhibited the proliferation of melanoma cells in vitro and exhibited concentration-dependent cell cycle arrest in G2/M phase. Mitotic arrest activated the intrinsic apoptotic machinery by increasing the cellular levels of cleaved poly ADP ribose polymerase (PARP) and fraction of sub-G1 cells. In vivo, LY293-loaded nanoparticles significantly inhibited the proliferation of highly aggressive metastasized melanoma in a syngeneic lung metastasis melanoma mouse model without toxicity to vital organs. In conclusion, we have designed a promising polymeric nanocarrier for systemic delivery of LY293 for treating metastatic melanoma while minimizing the toxicity associated with the administration of cosolvents.
Collapse
Affiliation(s)
- Vaibhav Mundra
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center (UNMC), 986025 Nebraska Medical Center, Omaha, NE, 68198-6025, USA
| | | | | | | | | | | |
Collapse
|
23
|
Phase I trial of biochemotherapy with cisplatin, temozolomide, and dose escalation of nab-paclitaxel combined with interleukin-2 and interferon-α in patients with metastatic melanoma. Melanoma Res 2015; 24:342-8. [PMID: 24743052 DOI: 10.1097/cmr.0000000000000062] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The primary objective of this study was to determine the safety, toxicity, and maximum tolerated dose of nanoparticle albumin-bound (nab)-paclitaxel as part of biochemotherapy for metastatic melanoma and to determine whether substituting nab-paclitaxel for less potent agents could increase response rates and duration. Treatment consisted of intravenous cisplatin (20 mg/m) on days 1-4, oral temozolomide (250 mg/m) on days 1-3, subcutaneous interferon-α (5×10 IU/m) on days 1-5, and continuous intravenous interleukin-2 (9×10 IU/m) for 96 h on days 1-4. A standard 3+3 dose escalation method was used; the nab-paclitaxel starting dose was 100 mg/m on day 1 and 70 mg/m on day 5. The treatment cycle was repeated every 3 weeks and toxicity was assessed weekly. Ten patients were enrolled. Dose-limiting toxicities included diarrhea, transaminasemia, and neutropenia. The maximum tolerated dose was not identified because the nab-paclitaxel dose on day 1 at the lowest planned dose (80 mg/m) caused dose-limiting toxicity in two of five patients. Of the nine patients who were evaluable for response, five had a partial response. The median time to disease progression was 5.30 months and the median overall survival was 8.73 months. Six patients developed central nervous system metastasis at a median of 5.33 months after treatment initiation. Biochemotherapy including nab-paclitaxel according to the doses and schedule regimen used in the present study has significant toxicity. Substituting dacarbazine with temozolomide did not prevent central nervous system metastasis in patients with metastatic melanoma.
Collapse
|
24
|
Porcelli L, Guida G, Quatrale AE, Cocco T, Sidella L, Maida I, Iacobazzi RM, Ferretta A, Stolfa DA, Strippoli S, Guida S, Tommasi S, Guida M, Azzariti A. Aurora kinase B inhibition reduces the proliferation of metastatic melanoma cells and enhances the response to chemotherapy. J Transl Med 2015; 13:26. [PMID: 25623468 PMCID: PMC4314759 DOI: 10.1186/s12967-015-0385-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 01/08/2015] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND The poor response to chemotherapy and the brief response to vemurafenib in metastatic melanoma patients, make the identification of new therapeutic approaches an urgent need. Interestingly the increased expression and activity of the Aurora kinase B during melanoma progression suggests it as a promising therapeutic target. METHODS The efficacy of the Aurora B kinase inhibitor barasertib-HQPA was evaluated in BRAF mutated cells, sensitive and made resistant to vemurafenib after chronic exposure to the drug, and in BRAF wild type cells. The drug effectiveness has been evaluated as cell growth inhibition, cell cycle progression and cell migration. In addition, cellular effectors of drug resistance and response were investigated. RESULTS The characterization of the effectors responsible for the resistance to vemurafenib evidenced the increased expression of MITF or the activation of Erk1/2 and p-38 kinases in the newly established cell lines with a phenotype resistant to vemurafenib. The sensitivity of cells to barasertib-HQPA was irrespective of BRAF mutational status. Barasertib-HQPA induced the mitotic catastrophe, ultimately causing apoptosis and necrosis of cells, inhibited cell migration and strongly affected the glycolytic metabolism of cells inducing the release of lactate. In association i) with vemurafenib the gain in effectiveness was found only in BRAF(V600K) cells while ii) with nab-paclitaxel, the combination was more effective than each drug alone in all cells. CONCLUSIONS These findings suggest barasertib as a new therapeutic agent and as enhancer of chemotherapy in metastatic melanoma treatment.
Collapse
Affiliation(s)
- Letizia Porcelli
- Clinical and Preclinical Pharmacology Laboratory, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Gabriella Guida
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari, P.zza Giulio Cesare, 70124, Bari, Italy.
| | - Anna E Quatrale
- Clinical and Preclinical Pharmacology Laboratory, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Tiziana Cocco
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari, P.zza Giulio Cesare, 70124, Bari, Italy.
| | - Letizia Sidella
- Clinical and Preclinical Pharmacology Laboratory, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Immacolata Maida
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari, P.zza Giulio Cesare, 70124, Bari, Italy.
| | - Rosa M Iacobazzi
- Clinical and Preclinical Pharmacology Laboratory, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Anna Ferretta
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari, P.zza Giulio Cesare, 70124, Bari, Italy.
| | - Diana A Stolfa
- Clinical and Preclinical Pharmacology Laboratory, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Sabino Strippoli
- Medical Oncology Department, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Stefania Guida
- Unit of Dermatology and Venereology, University of Bari, P.zza Giulio Cesare, 70124, Bari, Italy.
| | - Stefania Tommasi
- Molecular Genetics Laboratory, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Michele Guida
- Medical Oncology Department, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Amalia Azzariti
- Clinical and Preclinical Pharmacology Laboratory, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| |
Collapse
|
25
|
Sand KMK, Bern M, Nilsen J, Noordzij HT, Sandlie I, Andersen JT. Unraveling the Interaction between FcRn and Albumin: Opportunities for Design of Albumin-Based Therapeutics. Front Immunol 2015; 5:682. [PMID: 25674083 PMCID: PMC4306297 DOI: 10.3389/fimmu.2014.00682] [Citation(s) in RCA: 158] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 12/17/2014] [Indexed: 01/08/2023] Open
Abstract
The neonatal Fc receptor (FcRn) was first found to be responsible for transporting antibodies of the immunoglobulin G (IgG) class from the mother to the fetus or neonate as well as for protecting IgG from intracellular catabolism. However, it has now become apparent that the same receptor also binds albumin and plays a fundamental role in homeostatic regulation of both IgG and albumin, as FcRn is expressed in many different cell types and organs at diverse body sites. Thus, to gain a complete understanding of the biological function of each ligand, and also their distribution in the body, an in-depth characterization of how FcRn binds and regulates the transport of both ligands is necessary. Importantly, such knowledge is also relevant when developing new drugs, as IgG and albumin are increasingly utilized in therapy. This review discusses our current structural and biological understanding of the relationship between FcRn and its ligands, with a particular focus on albumin and design of albumin-based therapeutics.
Collapse
Affiliation(s)
- Kine Marita Knudsen Sand
- Department of Biosciences, Centre for Immune Regulation (CIR), University of Oslo , Oslo , Norway ; Department of Immunology, Centre for Immune Regulation (CIR), Oslo University Hospital Rikshospitalet , Oslo , Norway
| | - Malin Bern
- Department of Biosciences, Centre for Immune Regulation (CIR), University of Oslo , Oslo , Norway ; Department of Immunology, Centre for Immune Regulation (CIR), Oslo University Hospital Rikshospitalet , Oslo , Norway
| | - Jeannette Nilsen
- Department of Immunology, Centre for Immune Regulation (CIR), Oslo University Hospital Rikshospitalet , Oslo , Norway ; Institute of Clinical Medicine, University of Oslo , Oslo , Norway
| | - Hanna Theodora Noordzij
- Department of Biosciences, Centre for Immune Regulation (CIR), University of Oslo , Oslo , Norway ; Department of Immunology, Centre for Immune Regulation (CIR), Oslo University Hospital Rikshospitalet , Oslo , Norway
| | - Inger Sandlie
- Department of Biosciences, Centre for Immune Regulation (CIR), University of Oslo , Oslo , Norway ; Department of Immunology, Centre for Immune Regulation (CIR), Oslo University Hospital Rikshospitalet , Oslo , Norway
| | - Jan Terje Andersen
- Department of Immunology, Centre for Immune Regulation (CIR), Oslo University Hospital Rikshospitalet , Oslo , Norway
| |
Collapse
|
26
|
Megahed AI, Koon HB. What is the role of chemotherapy in the treatment of melanoma? Curr Treat Options Oncol 2015; 15:321-35. [PMID: 24599525 DOI: 10.1007/s11864-014-0277-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The approval of ipilimumab and inhibitors of the BRAF pathway for the treatment of melanoma has provided multiple therapeutic options for patients. Although these new agents improve survival compared with chemotherapy alone, the majority of patients will progress and will receive chemotherapy at some point in the course of their disease. Whether the clinical efficacy of chemotherapy can be improved by targeting resistance mechanisms is an area of active investigation. In addition, chemotherapy may be of use modulating the efficacy of the newer agents.
Collapse
Affiliation(s)
- Ahmed I Megahed
- University Hospitals Case Medical Center, Cleveland, OH, 44106, USA
| | | |
Collapse
|
27
|
Garrisi VM, Strippoli S, De Summa S, Pinto R, Perrone A, Guida G, Azzariti A, Guida M, Stefania T. Proteomic profile and in silico analysis in metastatic melanoma with and without BRAF mutation. PLoS One 2014; 9:e112025. [PMID: 25437182 PMCID: PMC4249853 DOI: 10.1371/journal.pone.0112025] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 10/11/2014] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION Selective inhibitors of BRAF, vemurafenib and dabrafenib are the standard of care for metastatic melanoma patients with BRAF V600, while chemotherapy continued to be widely used in BRAF wild type patients. MATERIALS AND METHODS In order to discover novel candidate biomarkers predictive to treatment, serum of 39 metastatic melanoma vemurafenib (n = 19) or chemotherapy (n = 20) treated patients at baseline, at disease control and at progression, were analyzed using SELDI-TOF technology. In silico analysis was used to identify more significant peaks. RESULTS In patients with different BRAF status, we found 5 peptides significantly deregulated, with the down-regulation of the m/z 9176 peak strongly associated with BRAF mutation. At baseline as predictive biomarkers we identified 2 peptides - m/z 6411, 4075 - as significantly up-regulated in responders to chemotherapy and 4 peaks - m/z 5900, 12544, 49124 and 11724 - significantly up-regulated in longer vs shorter responders to vemurafenib. After response, 3 peptides (m/z 4658, 18639, and 9307) resulted significantly down regulated while 3 peptides m/z 9292, 7765 and 9176 appeared up-regulated respectively in chemotherapy and vemurafenib responder patients. In vemurafenib treated patients, 16 peaks appeared deregulated at progression compared to baseline time. In silico analysis identified proteins involved in invasiveness (SLAIN1) and resistance (ABCC12) as well as in the pathway of detoxification (NQO1) and apoptosis (RBM10, TOX3, MTEFD1, TSPO2). Proteins associated with the modulation of neuronal plasticity (RIN1) and regulatory activity factors of gene transcription (KLF17, ZBTB44) were also highlighted. CONCLUSION Our exploratory study highlighted some factors that deserve to be further investigated in order to provide a framework for improving melanoma treatment management through the development of biomarkers which could act as the strongest surrogates of the key biological events in stage IV melanoma.
Collapse
Affiliation(s)
- Vito Michele Garrisi
- National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Sabino Strippoli
- National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Simona De Summa
- National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Rosamaria Pinto
- National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Antonella Perrone
- National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Gabriella Guida
- Dept. of Basic Medical Sciences, Faculty of Medicine and Surgery, School of Medicine, University of Bari, Bari, Italy
| | - Amalia Azzariti
- National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Michele Guida
- National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Tommasi Stefania
- National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Bari, Italy
- * E-mail:
| |
Collapse
|
28
|
Lee RJ, Ul-Ain-Tariq N, Fusi A, Bowyer S, Lorigan P. The role of chemotherapy in the modern management of melanoma. Melanoma Manag 2014; 1:173-184. [PMID: 30190822 PMCID: PMC6094611 DOI: 10.2217/mmt.14.20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The last 4 years have seen dramatic changes in the treatment of advanced melanoma, largely based on advances in targeted therapy and immunotherapy. This article examines the role of chemotherapy in the modern management of melanoma. We examine the evidence for promising new agents and discuss their position in the sequencing of treatment options for patients with advanced disease. In addition, we discuss the combination of chemotherapy with targeted treatments and immune therapies. Finally, we discuss future areas of research for ensuring that we maximize the potential of all agents available to us and identify new, effective treatments.
Collapse
Affiliation(s)
- Rebecca Jane Lee
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK
- The University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - Noor Ul-Ain-Tariq
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK
| | - Alberto Fusi
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK
| | - Samantha Bowyer
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK
| | - Paul Lorigan
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK
- The University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| |
Collapse
|
29
|
Drug delivery nanoparticles in skin cancers. BIOMED RESEARCH INTERNATIONAL 2014; 2014:895986. [PMID: 25101298 PMCID: PMC4102061 DOI: 10.1155/2014/895986] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 04/28/2014] [Indexed: 12/17/2022]
Abstract
Nanotechnology involves the engineering of functional systems at nanoscale, thus being attractive for disciplines ranging from materials science to biomedicine. One of the most active research areas of the nanotechnology is nanomedicine, which applies nanotechnology to highly specific medical interventions for prevention, diagnosis, and treatment of diseases, including cancer disease. Over the past two decades, the rapid developments in nanotechnology have allowed the incorporation of multiple therapeutic, sensing, and targeting agents into nanoparticles, for detection, prevention, and treatment of cancer diseases. Nanoparticles offer many advantages as drug carrier systems since they can improve the solubility of poorly water-soluble drugs, modify pharmacokinetics, increase drug half-life by reducing immunogenicity, improve bioavailability, and diminish drug metabolism. They can also enable a tunable release of therapeutic compounds and the simultaneous delivery of two or more drugs for combination therapy. In this review, we discuss the recent advances in the use of different types of nanoparticles for systemic and topical drug delivery in the treatment of skin cancer. In particular, the progress in the treatment with nanocarriers of basal cell carcinoma, squamous cell carcinoma, and melanoma has been reported.
Collapse
|
30
|
Shah DJ, Dronca RS. Latest advances in chemotherapeutic, targeted, and immune approaches in the treatment of metastatic melanoma. Mayo Clin Proc 2014; 89:504-19. [PMID: 24684873 PMCID: PMC4034544 DOI: 10.1016/j.mayocp.2014.02.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 02/03/2014] [Accepted: 02/05/2014] [Indexed: 12/28/2022]
Abstract
Melanoma is the most dangerous form of skin cancer owing to its metastatic potential and is an important public health concern. The melanoma incidence has been increasing worldwide. Although potentially curable when diagnosed early, metastatic melanoma carries a poor prognosis. Until recently, systemic therapy for metastatic melanoma was ineffective, but the recent successes in the development of new therapies for metastatic melanoma, such as mitogen-activated protein kinase (MAPK) pathway inhibitors, anti-cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4), and programmed cell death protein 1 (PD-1)/programmed cell death ligand 1 (PD-L1) pathway blocking antibodies, as well as combination strategies of cytotoxic chemotherapy and inhibitors of angiogenesis, have all yielded promising results, changing the continually evolving landscape of therapeutic options for patients with this disease. The aim of this review was to summarize the evolution of and recent advances in the treatment of metastatic melanoma. Therefore, we conducted a comprehensive PubMed search between January 1, 1960, and February 1, 2014, using the search term melanoma or metastatic melanoma combined with terms such as chemotherapy, immunotherapy, CTLA-4, PD-1, PD-L1, adoptive T cell, targeted therapy, MAPK, molecular biology, and survival.
Collapse
Affiliation(s)
- Darshil J Shah
- Department of Internal Medicine, Oakland University William Beaumont School of Medicine, Beaumont Health System, Royal Oak, MI.
| | | |
Collapse
|
31
|
Abstract
The recent past has witnessed unprecedented clinical progress in the treatment of advanced malignant melanoma through targeting of mutant BRAF in approximately 50% of patients and immune check point blockade in all patients. As has been well documented, responses to targeted therapy are of limited duration, and rates of clinical benefit to immunotherapy are modest. Given these factors, palliation of patients with chemotherapy remains an essential aspect of melanoma oncology. Many chemotherapeutics (and combinations with other agents, such as immunotherapy) have been evaluated in melanoma, although no chemotherapy regimen has been documented to provide an overall survival benefit in a prospective, randomized, well-controlled phase III study. We provide an overview of the development of the most common chemotherapy regimens for melanoma, discuss the clinical trial evidence supporting and contrasting them, and highlight appropriate clinical situations in which they might be used. We also discuss the future of chemotherapy for melanoma, noting the potential for combinations of chemotherapy with either targeted or immunotherapeutic agents.
Collapse
Affiliation(s)
- Jason J Luke
- Department of Medicine, Melanoma and Sarcoma Service, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | | |
Collapse
|
32
|
Chen J, Shao R, Zhang XD, Chen C. Applications of nanotechnology for melanoma treatment, diagnosis, and theranostics. Int J Nanomedicine 2013; 8:2677-88. [PMID: 23926430 PMCID: PMC3728269 DOI: 10.2147/ijn.s45429] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Melanoma is the most aggressive type of skin cancer and has very high rates of mortality. An early stage melanoma can be surgically removed, with a survival rate of 99%. However, metastasized melanoma is difficult to cure. The 5-year survival rates for patients with metastasized melanoma are still below 20%. Metastasized melanoma is currently treated by chemotherapy, targeted therapy, immunotherapy and radiotherapy. The outcome of most of the current therapies is far from optimistic. Although melanoma patients with a mutation in the oncogene v-Raf murine sarcoma viral oncogene homolog B1 (BRAF) have an initially higher positive response rate to targeted therapy, the majority develop acquired drug resistance after 6 months of the therapy. To increase treatment efficacy, early diagnosis, more potent pharmacological agents, and more effective delivery systems are urgently needed. Nanotechnology has been extensively studied for melanoma treatment and diagnosis, to decrease drug resistance, increase therapeutic efficacy, and reduce side effects. In this review, we summarize the recent progress on the development of various nanoparticles for melanoma treatment and diagnosis. Several common nanoparticles, including liposome, polymersomes, dendrimers, carbon-based nanoparticles, and human albumin, have been used to deliver chemotherapeutic agents, and small interfering ribonucleic acids (siRNAs) against signaling molecules have also been tested for the treatment of melanoma. Indeed, several nanoparticle-delivered drugs have been approved by the US Food and Drug Administration and are currently in clinical trials. The application of nanoparticles could produce side effects, which will need to be reduced so that nanoparticle-delivered drugs can be safely applied in the clinical setting.
Collapse
Affiliation(s)
- Jiezhong Chen
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia.
| | | | | | | |
Collapse
|
33
|
Batus M, Waheed S, Ruby C, Petersen L, Bines SD, Kaufman HL. Optimal management of metastatic melanoma: current strategies and future directions. Am J Clin Dermatol 2013; 14:179-94. [PMID: 23677693 PMCID: PMC3913474 DOI: 10.1007/s40257-013-0025-9] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Melanoma is increasing in incidence and remains a major public health threat. Although the disease may be curable when identified early, advanced melanoma is characterized by widespread metastatic disease and a median survival of less than 10 months. In recent years, however, major advances in our understanding of the molecular nature of melanoma and the interaction of melanoma cells with the immune system have resulted in several new therapeutic strategies that are showing significant clinical benefit. Current therapeutic approaches include surgical resection of metastatic disease, chemotherapy, immunotherapy, and targeted therapy. Dacarbazine, interleukin-2, ipilimumab, and vemurafenib are now approved for the treatment of advanced melanoma. In addition, new combination chemotherapy regimens, monoclonal antibodies blocking the programmed death-1 (PD-1)/PD-ligand 1 pathway, and targeted therapy against CKIT, mitogen-activated protein/extracellular signal-regulated kinase (MEK), and other putative signaling pathways in melanoma are beginning to show promise in early-phase clinical trials. Further research on these modalities alone and in combination will likely be the focus of future clinical investigation and may impact the outcomes for patients with advanced melanoma.
Collapse
Affiliation(s)
- Marta Batus
- Rush University Melanoma Program and Departments of Medicine, General Surgery and Immunology and Microbiology, Rush University Medical Center, 1725 W. Harrison Street, Room 845, Chicago, IL 60612, USA
| | | | | | | | | | | |
Collapse
|
34
|
Nanoparticle albumin bound Paclitaxel in the treatment of human cancer: nanodelivery reaches prime-time? JOURNAL OF DRUG DELIVERY 2013; 2013:905091. [PMID: 23738077 PMCID: PMC3659516 DOI: 10.1155/2013/905091] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 03/05/2013] [Indexed: 01/17/2023]
Abstract
Nanoparticle albumin bound paclitaxel (nab-paclitaxel) represents the first nanotechnology-based drug in cancer treatment. We discuss the development of this innovative compound and report the recent changing-practice results in breast and pancreatic cancer. A ground-breaking finding is the demonstration that nab-paclitaxel can not only enhance the activity and reduce the toxicity of chromophore-diluted compound, but also exert activity in diseases considered refractory to taxane-based treatment. This is the first clinical demonstration of major activity of nanotechnologically modified drugs in the treatment of human neoplasms.
Collapse
|
35
|
A phase I study of hepatic arterial infusion of nab-paclitaxel in combination with intravenous gemcitabine and bevacizumab for patients with advanced cancers and predominant liver metastases. Cancer Chemother Pharmacol 2013; 71:955-63. [PMID: 23377373 DOI: 10.1007/s00280-013-2088-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 01/12/2013] [Indexed: 12/29/2022]
Abstract
PURPOSE We conducted a phase I clinical trial for patients with advanced cancer and predominant liver disease. METHODS Patients were treated with HAI nab-paclitaxel (120-210 mg/m(2); day 1); intravenous bevacizumab (10 mg/kg; day 1); and intravenous gemcitabine (600-800 mg/m(2); days 1 and 8). A conventional "3 + 3" study design was used. RESULTS Fifty patients with advanced cancer and predominant liver metastases were treated (median age, 58 years; 27 women, 23 men; median number of prior therapies, 3 [range 0-12]). The most common cancers were breast (n = 9) and pancreatic (n = 9). Overall, 264 cycles were administered (median/patient, 4; range 1-17). No dose-limiting toxicities were noted during the escalation phase. On dose level 4, 3 patients were unable to receive gemcitabine on day 8 because of severe thrombocytopenia. Dose level 3 was selected as the maximum-tolerated dose (HAI nab-paclitaxel 180 mg/m(2) and intravenous gemcitabine 800 mg/m(2) and bevacizumab 10 mg/kg); 32 patients were treated in the expansion phase. The most common treatment-related toxicities were thrombocytopenia (n = 17), neutropenia (n = 10), and fatigue (n = 12). Of 46 patients evaluable for response, 9 (20 %) had a partial response (PR) and 9 (20 %) had stable disease for ≥6 months. The median overall survival duration was 7.0 months (95 % CI: 4, 22 months), and the median progression-free survival duration was 4.2 months (95 % CI: 2.7, 8.6 months). CONCLUSIONS HAI nab-paclitaxel in combination with gemcitabine and bevacizumab was well tolerated and had antitumor activity in selected patients with advanced cancer and liver metastases.
Collapse
|
36
|
Velho TR. Metastatic melanoma - a review of current and future drugs. Drugs Context 2012; 2012:212242. [PMID: 24432031 PMCID: PMC3885142 DOI: 10.7573/dic.212242] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 08/20/2012] [Indexed: 12/21/2022] Open
Abstract
Background: Melanoma is one of the most aggressive cancers, and it is estimated that 76,250 men and women will be diagnosed with melanoma of the skin in the USA in 2012. Over the last few decades many drugs have been developed but only in 2011 have new drugs demonstrated an impact on survival in metastatic melanoma. Methods: A systematic search of literature was conducted, and studies providing data on the effectiveness of current and/or future drugs used in the treatment of metastatic melanoma were selected for review. This review discusses the advantages and limitations of these agents, evaluating past, current and future clinical trials designed to overcome such limitations. Results: To date, there are four drugs approved by the Food and Drug Administration for melanoma (dacarbazine, interleukin-2, ipilimumab and vemurafenib). Despite efforts to develop new drugs, few of them have demonstrated any clinical benefits. Approved in 1975, dacarbazine remains the gold standard in chemotherapy, although ipilimumab and vemurafenib have raised many hopes in the last few years. Combining dacarbazine or other chemotherapy agents with new pharmacological agents may be a new way to achieve better clinical responses in patients with metastatic melanoma. Discussion: Advances in the molecular knowledge of melanoma have led to major improvements in the treatment of patients with metastatic melanoma, providing new targets and insights. However, heterogeneity amongst study populations, different approaches to treatment and the different melanoma types and localisations included in the trials makes their comparison difficult. New studies focusing on drugs developed in recent decades are warranted.
Collapse
|
37
|
Oblimersen in combination with temozolomide and albumin-bound paclitaxel in patients with advanced melanoma: a phase I trial. Cancer Chemother Pharmacol 2012; 71:183-91. [PMID: 23064957 DOI: 10.1007/s00280-012-1995-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 09/29/2012] [Indexed: 10/27/2022]
Abstract
PURPOSE The combination of oblimersen, a bcl-2 antisense oligonucleotide, and dacarbazine lead to superior progression-free survival in advanced melanoma patients. Albumin-bound paclitaxel (nab-paclitaxel) has single-agent activity in melanoma. METHODS In a phase I trial, chemotherapy-naïve patients with metastatic melanoma and normal LDH levels were enrolled on 3 cohorts. The treatment regimen consisted of 56-day cycles of oblimersen (7 mg/kg/day continuous IV infusion on day 1-7 and 22-28 in cohort 1 and 2; 900 mg fixed dose, twice weekly in weeks 1-2, 4-5 for cohort 3), temozolomide (75 mg/m(2), days 1-42), and nab-paclitaxel (175 mg/m(2) in cohort 1 and 3, 260 mg/m(2) in cohort 2 on day 7 and 28). Apoptosis markers were tested in pre- and post-treatment specimens of a subset of patients. RESULTS Six grade 3 events (neutropenia, renal insufficiency, hyponatremia, elevated creatinine, allergic reaction, and neuropathy) and 2 grade 4 events (neutropenia and thrombocytopenia) were seen in 32 patients. The objective response rate was 40.6% (2 complete responses and 11 partial responses) and 11 patients had stable disease, for a disease control rate of 75%. CONCLUSIONS The combination of oblimersen, temozolomide, and nab-paclitaxel was well tolerated and demonstrated encouraging activity in patients with advanced melanoma.
Collapse
|
38
|
Espinosa E, Berrocal A, López Martín JA, González Cao M, Cerezuela P, Mayordomo JI, Martín Algarra S. Advances in cutaneous melanoma. Clin Transl Oncol 2012; 14:325-32. [PMID: 22551537 DOI: 10.1007/s12094-012-0804-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
After several decades of slow progress in the field of melanoma, significant advances have been reported in recent years. These include a better understanding of the molecular biology of the tumour, a new staging classification system, insights into the patterns of relapse in early stage, and new drugs for the treatment of advanced disease. Ipilimumab and vemurafenib have just been approved and provide a survival benefit in stage IV. Both compounds are under evaluation in the adjuvant setting, where interferon remains the only drug with proven efficacy. Further investigation is required to treat patients with primary or secondary resistance to new drugs.
Collapse
|
39
|
Von Hoff DD, Ramanathan RK, Borad MJ, Laheru DA, Smith LS, Wood TE, Korn RL, Desai N, Trieu V, Iglesias JL, Zhang H, Soon-Shiong P, Shi T, Rajeshkumar NV, Maitra A, Hidalgo M. Gemcitabine plus nab-paclitaxel is an active regimen in patients with advanced pancreatic cancer: a phase I/II trial. J Clin Oncol 2011; 29:4548-54. [PMID: 21969517 DOI: 10.1200/jco.2011.36.5742] [Citation(s) in RCA: 860] [Impact Index Per Article: 61.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
PURPOSE The trial objectives were to identify the maximum-tolerated dose (MTD) of first-line gemcitabine plus nab-paclitaxel in metastatic pancreatic adenocarcinoma and to provide efficacy and safety data. Additional objectives were to evaluate positron emission tomography (PET) scan response, secreted protein acidic and rich in cysteine (SPARC), and CA19-9 levels in relation to efficacy. Subsequent preclinical studies investigated the changes involving the pancreatic stroma and drug uptake. PATIENTS AND METHODS Patients with previously untreated advanced pancreatic cancer were treated with 100, 125, or 150 mg/m(2) nab-paclitaxel followed by gemcitabine 1,000 mg/m(2) on days 1, 8, and 15 every 28 days. In the preclinical study, mice were implanted with human pancreatic cancers and treated with study agents. RESULTS A total of 20, 44, and three patients received nab-paclitaxel at 100, 125, and 150 mg/m(2), respectively. The MTD was 1,000 mg/m(2) of gemcitabine plus 125 mg/m(2) of nab-paclitaxel once a week for 3 weeks, every 28 days. Dose-limiting toxicities were sepsis and neutropenia. At the MTD, the response rate was 48%, with 12.2 median months of overall survival (OS) and 48% 1-year survival. Improved OS was observed in patients who had a complete metabolic response on [(18)F]fluorodeoxyglucose PET. Decreases in CA19-9 levels were correlated with increased response rate, progression-free survival, and OS. SPARC in the stroma, but not in the tumor, was correlated with improved survival. In mice with human pancreatic cancer xenografts, nab-paclitaxel alone and in combination with gemcitabine depleted the desmoplastic stroma. The intratumoral concentration of gemcitabine was increased by 2.8-fold in mice receiving nab-paclitaxel plus gemcitabine versus those receiving gemcitabine alone. CONCLUSION The regimen of nab-paclitaxel plus gemcitabine has tolerable adverse effects with substantial antitumor activity, warranting phase III evaluation.
Collapse
Affiliation(s)
- Daniel D Von Hoff
- TGen/Virginia G Piper Cancer Ctr, 445 N Fifth St, Suite 600, Phoenix, AZ 85004, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Whole recombinant yeast vaccine induces antitumor immunity and improves survival in a genetically engineered mouse model of melanoma. Gene Ther 2011; 18:827-34. [PMID: 21390072 DOI: 10.1038/gt.2011.28] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Malignant melanoma is one of the deadliest forms of skin cancer and its incidence is expected to rise over the next two decades. At present, there are no effective therapies for advanced melanoma. We have previously shown that administration of whole recombinant yeast expressing human MART-1 (hMART-IT) induces protective antimelanoma immunity in a B16F10 transplantable mouse model. In this study, we examine the effectiveness of the hMART-IT vaccine in a congenic strain of genetically engineered mouse model of melanoma, which recapitulates both the underlying genetics and the proper tumor microenvironment of naturally occurring melanoma. Subcutaneous administration of hMART-IT induced cytotoxicity against melanoma cells and antigen-specific production of Th1-specific cytokines by splenocytes. Weekly administration of hMART-IT significantly delayed the development of melanoma and prolonged the survival of mice compared with controls. Although histological analysis demonstrated diffuse infiltration of CD4(+) T cells and CD8(+) T cells, no reduction of regulatory T cells was observed, suggesting that hMART-IT cannot prevent immunotolerance in the tumor microenvironment. This study provides a proof of concept that genetically engineered mouse models lend valuable insights into immunotherapeutics being tested in the preclinical setting.
Collapse
|
41
|
Kottschade LA, Suman VJ, Amatruda T, McWilliams RR, Mattar BI, Nikcevich DA, Behrens R, Fitch TR, Jaslowski AJ, Markovic SN. A phase II trial of nab-paclitaxel (ABI-007) and carboplatin in patients with unresectable stage IV melanoma : a North Central Cancer Treatment Group Study, N057E(1). Cancer 2010; 117:1704-10. [PMID: 21472717 DOI: 10.1002/cncr.25659] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2010] [Revised: 08/17/2010] [Accepted: 08/18/2010] [Indexed: 11/06/2022]
Abstract
BACKGROUND There is increasing evidence that paclitaxel and carboplatin are clinically active in the treatment of metastatic melanoma (MM). ABI-007 is an albumin-bound formulation of paclitaxel that has demonstrated single-agent activity against metastatic melanoma. METHODS A parallel phase II trial was conducted in patients with unresectable stage IV melanoma who were either chemotherapy naive (CN) or previously treated (PT). The treatment regimen consisted of ABI-007 (100 mg/m(2) ) and carboplatin area under the curve (AUC2) administered on days 1, 8, and 15 every 28 days. The primary aim of this study was objective response rate (RECIST). RESULTS Seventy-six patients (41 CN and 35 PT) were enrolled between November 2006 and July 2007. Three patients withdrew consent prior to starting treatment. The median number of treatment cycles was 4. There were 10 (25.6%) responses (1 complete response [CR] and 9 partial responses [PRs]) in the CN cohort (90% CI, 16.7%-42.3%) and 3 (8.8%) responses (3 PRs) in the PT cohort (90% CI, 2.5%-21.3%). Median progression-free survival was 4.5 months in the CN cohort and 4.1 months in the PT cohort. Median overall survival (OS) was 11.1 months in the CN group and 10.9 months in the PT group. Severe toxicities in both groups (Common Terminology Criteria for Adverse Effects v.3.0 ≥grade 3) included neutropenia, thrombocytopenia, neurosensory problems, fatigue, nausea, and vomiting. CONCLUSIONS The weekly combination of ABI-007 and carboplatin appears to be moderately well tolerated, with promising clinical activity as therapy in patients who are chemotherapy naive and with modest antitumor activity in those previously treated.
Collapse
|
42
|
Sarin H. Overcoming the challenges in the effective delivery of chemotherapies to CNS solid tumors. Ther Deliv 2010; 1:289-305. [PMID: 22163071 PMCID: PMC3234205 DOI: 10.4155/tde.10.22] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Locoregional therapies, such as surgery and intratumoral chemotherapy, do not effectively treat infiltrative primary CNS solid tumors and multifocal metastatic solid tumor disease of the CNS. It also remains a challenge to treat such CNS malignant solid tumor disease with systemic chemotherapies, although these lipid-soluble small-molecule drugs demonstrate potent cytotoxicity in vitro. Even in the setting of a 'normalized' tumor microenvironment, small-molecule drugs do not accumulate to effective concentrations in the vast majority of tumor cells, which is due to the fact that small-molecule drugs have short blood half-lives. It has been recently shown that drug-conjugated spherical lipid-insoluble nanoparticles within the 7-10 nm size range can deliver therapeutic concentrations of drug fraction directly into individual tumor cells following systemic administration, since these functionalized particles maintain peak blood concentrations for several hours and are smaller than the physiologic upper limit of pore size in the VEGF-derived blood capillaries of solid tumors, which is approximately 12 nm. In this article, the physiologic and ultrastructural basis of this novel translational approach for the treatment of CNS, as well as non-CNS, solid cancers is reviewed.
Collapse
Affiliation(s)
- Hemant Sarin
- National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, USA.
| |
Collapse
|