1
|
Joshi DC, Sharma A, Prasad S, Singh K, Kumar M, Sherawat K, Tuli HS, Gupta M. Novel therapeutic agents in clinical trials: emerging approaches in cancer therapy. Discov Oncol 2024; 15:342. [PMID: 39127974 PMCID: PMC11317456 DOI: 10.1007/s12672-024-01195-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Novel therapeutic agents in clinical trials offer a paradigm shift in the approach to battling this prevalent and destructive disease, and the area of cancer therapy is on the precipice of a trans formative revolution. Despite the importance of tried-and-true cancer treatments like surgery, radiation, and chemotherapy, the disease continues to evolve and adapt, making new, more potent methods necessary. The field of cancer therapy is currently witnessing the emergence of a wide range of innovative approaches. Immunotherapy, including checkpoint inhibitors, CAR-T cell treatment, and cancer vaccines, utilizes the host's immune system to selectively target and eradicate malignant cells while minimizing harm to normal tissue. The development of targeted medicines like kinase inhibitors and monoclonal antibodies has allowed for more targeted and less harmful approaches to treating cancer. With the help of genomics and molecular profiling, "precision medicine" customizes therapies to each patient's unique genetic makeup to maximize therapeutic efficacy while minimizing unwanted side effects. Epigenetic therapies, metabolic interventions, radio-pharmaceuticals, and an increasing emphasis on combination therapy with synergistic effects further broaden the therapeutic landscape. Multiple-stage clinical trials are essential for determining the safety and efficacy of these novel drugs, allowing patients to gain access to novel treatments while also furthering scientific understanding. The future of cancer therapy is rife with promise, as the integration of artificial intelligence and big data has the potential to revolutionize early detection and prevention. Collaboration among researchers, and healthcare providers, and the active involvement of patients remain the bedrock of the ongoing battle against cancer. In conclusion, the dynamic and evolving landscape of cancer therapy provides hope for improved treatment outcomes, emphasizing a patient-centered, data-driven, and ethically grounded approach as we collectively strive towards a cancer-free world.
Collapse
Affiliation(s)
- Deepak Chandra Joshi
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandar Sindri, Dist., Ajmer, Rajasthan, India.
| | - Anurag Sharma
- Invertis Institute of Pharmacy, Invertis University Bareilly Uttar Pradesh, Bareilly, India
| | - Sonima Prasad
- Chandigarh University, Ludhiana-Chandigarh State Highway, Gharuan, Mohali, Punjab, 140413, India
| | - Karishma Singh
- Institute of Pharmaceutical Sciences, Faculty of Engineering and Technology, University of Lucknow, Lucknow, India
| | - Mayank Kumar
- Himalayan Institute of Pharmacy, Road, Near Suketi Fossil Park, Kala Amb, Hamidpur, Himachal Pradesh, India
| | - Kajal Sherawat
- Meerut Institute of Technology, Meerut, Uttar Pradesh, India
| | - Hardeep Singh Tuli
- Department of Bio-Sciences & Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, India
| | - Madhu Gupta
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, India.
| |
Collapse
|
2
|
Fathima T, Arumugam P, AS SG, Priyadharsini JV. Decoding the Genetic Alterations in Genes of DNMT Family (DNA Methyl-Transferase) and their Association with Head and Neck Squamous Cell Carcinoma. Asian Pac J Cancer Prev 2020; 21:3605-3612. [PMID: 33369458 PMCID: PMC8046301 DOI: 10.31557/apjcp.2020.21.12.3605] [Citation(s) in RCA: 4] [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] [Received: 08/03/2020] [Accepted: 11/26/2020] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE Epigenetic modifications are gaining focus due to their indirect association with tumorigenesis. DNA methylation plays a prime role in regulation of gene expression. Any aberrations in this gene family may lead to chromosomal instability and increased magnitude of tumour progression. In line with the above fact, the present study has been designed to identify genetic alterations in the genes of the DNMT (DNA methyl-transferase) family among head and neck squamous cell carcinoma patients (HNSCC). METHODS The present study follows an observational design employing computational tools for analysis. The TCGA-Firehose Legacy data was assessed using the cBioportal database. The dataset comprised of 530 samples from HNSCC patients which were assessed for genetic alterations in the DNMT family. Furthermore, the protein stability analysis and pathogenicity of the mutations were assessed using I-Mutant Suite and PROVEAN tools. RESULTS Almost all genes of the DNMT family harboured gene amplification. The TRDMT1 and DNMT3L genes showed deep deletions. Apart from these several non-synonymous, truncating and splice-site mutations were also documented. Protein stability and pathogenicity analysis revealed that majority of the mutations were found to decrease the stability and impose pathogenicity. Upon probing for reported mutations using gnomAD database, around six reference single nucleotide polymorphisms were identified which were found to exhibit a minor allele frequency less than 0.01. CONCLUSIONS Screening of an exhaustive collection of patient's samples could provide immense knowledge about the disease pathogenesis and identification of therapeutic leads. The variants identified in the present study could be used as diagnostic markers. However, further experimental analysis through genotyping assay is warranted to validate the present findings. .
Collapse
Affiliation(s)
- Tahreem Fathima
- Saveetha Dental College, Velappanchavadi, Poonamallee High Road, India.
| | - Paramasivam Arumugam
- Dental Research Cell, Saveetha Dental College, Poonamallee High Road, Chennai-77, India.
| | - Smiline Girija AS
- Department of Microbiology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, India.
| | - J Vijayashree Priyadharsini
- Biomedical Research Unit and Laboratory Animal Centre - Dental Research Cell, Saveetha Dental College and Hospitals, Chennai-77, India.
| |
Collapse
|
3
|
Mascaro-Cordeiro B, Oliveira ID, Tesser-Gamba F, Pavon LF, Saba-Silva N, Cavalheiro S, Dastoli P, Toledo SRC. Valproic acid treatment response in vitro is determined by TP53 status in medulloblastoma. Childs Nerv Syst 2018; 34:1497-1509. [PMID: 29785653 DOI: 10.1007/s00381-018-3817-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 04/25/2018] [Indexed: 12/18/2022]
Abstract
PURPOSE Histone deacetylate inhibitors (HDACi), as valproic acid (VA), have been reported to enhance efficacy and to prevent drug resistance in some tumors, including medulloblastoma (MB). In the present study, we investigated VA role, combined to cisplatin (CDDP) in cell viability and gene expression of MB cell lines. METHODS Dose-response curve determined IC50 values for each treatment: (1) VA single, (2) CDDP single, and (3) VA and CDDP combined. Cytotoxicity and flow cytometry evaluated cell viability after exposure to treatments. Quantitative PCR evaluated gene expression levels of AKT, CTNNB1, GLI1, KDM6A, KDM6B, NOTCH2, PTCH1, and TERT, before and after treatment. Besides, we performed next-generation sequencing (NGS) for PTCH1, TERT, and TP53 genes. RESULTS The most effective treatment to reduce viability was combined for D283MED and ONS-76; and CDDP single for DAOY cells (p < 0.0001). TERT, GLI1, and AKT genes were overexpressed after treatments with VA. D283MED and ONS-76 cells presented variants in TERT and PTCH1, respectively and DAOY cell line presented a TP53 mutation. CONCLUSIONS MB tumors belonging to SHH molecular subgroup, with TP53MUT, would be the ones that present high risk in relation to VA use during the treatment, while TP53WT MBs can benefit from VA therapy, both SHH and groups 3 and 4. Our study shows a new perspective about VA action in medulloblastoma cells, raising the possibility that VA may act in different patterns. According to the genetic background of MB cell, VA can stimulate cell cycle arrest and apoptosis or induce resistance to treatment via signaling pathways activation.
Collapse
Affiliation(s)
- Bruna Mascaro-Cordeiro
- Pediatrics Oncology Institute-GRAACC (Grupo de Apoio ao Adolescente e à Criança com Câncer) /UNIFESP (Federal University of Sao Paulo), Rua Botucatu, 743, Floor 8 - Genetics Laboratory, Vila Clementino, São Paulo, SP, Zip Code 04023-062, Brazil.,Department of Morphology and Genetics, Division of Genetics, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Indhira Dias Oliveira
- Pediatrics Oncology Institute-GRAACC (Grupo de Apoio ao Adolescente e à Criança com Câncer) /UNIFESP (Federal University of Sao Paulo), Rua Botucatu, 743, Floor 8 - Genetics Laboratory, Vila Clementino, São Paulo, SP, Zip Code 04023-062, Brazil
| | - Francine Tesser-Gamba
- Pediatrics Oncology Institute-GRAACC (Grupo de Apoio ao Adolescente e à Criança com Câncer) /UNIFESP (Federal University of Sao Paulo), Rua Botucatu, 743, Floor 8 - Genetics Laboratory, Vila Clementino, São Paulo, SP, Zip Code 04023-062, Brazil
| | - Lorena Favaro Pavon
- Departament of Neurology, Federal University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Nasjla Saba-Silva
- Pediatrics Oncology Institute-GRAACC (Grupo de Apoio ao Adolescente e à Criança com Câncer) /UNIFESP (Federal University of Sao Paulo), Rua Botucatu, 743, Floor 8 - Genetics Laboratory, Vila Clementino, São Paulo, SP, Zip Code 04023-062, Brazil
| | - Sergio Cavalheiro
- Pediatrics Oncology Institute-GRAACC (Grupo de Apoio ao Adolescente e à Criança com Câncer) /UNIFESP (Federal University of Sao Paulo), Rua Botucatu, 743, Floor 8 - Genetics Laboratory, Vila Clementino, São Paulo, SP, Zip Code 04023-062, Brazil.,Departament of Neurology, Federal University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Patrícia Dastoli
- Pediatrics Oncology Institute-GRAACC (Grupo de Apoio ao Adolescente e à Criança com Câncer) /UNIFESP (Federal University of Sao Paulo), Rua Botucatu, 743, Floor 8 - Genetics Laboratory, Vila Clementino, São Paulo, SP, Zip Code 04023-062, Brazil
| | - Silvia Regina Caminada Toledo
- Pediatrics Oncology Institute-GRAACC (Grupo de Apoio ao Adolescente e à Criança com Câncer) /UNIFESP (Federal University of Sao Paulo), Rua Botucatu, 743, Floor 8 - Genetics Laboratory, Vila Clementino, São Paulo, SP, Zip Code 04023-062, Brazil. .,Department of Morphology and Genetics, Division of Genetics, Federal University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
4
|
Ling Y, Feng J, Luo L, Guo J, Peng Y, Wang T, Ge X, Xu Q, Wang X, Dai H, Zhang Y. Design and Synthesis of C3-Substituted β-Carboline-Based Histone Deacetylase Inhibitors with Potent Antitumor Activities. ChemMedChem 2017; 12:646-651. [PMID: 28425177 DOI: 10.1002/cmdc.201700133] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 04/04/2017] [Indexed: 12/11/2022]
Abstract
A series of hydroxamic acid histone deacetylase (HDAC) inhibitors in which the β-carboline motif has been incorporated were designed and synthesized. The effect of substitution at the C3 amide on HDAC inhibition and antiproliferative activities was investigated. Most of these compounds were found to display significant HDAC inhibitory effects and good antiproliferative activity, with IC50 values in the low-micromolar range. In particular, the HDAC inhibition IC50 value of N-(2-(dimethylamino)ethyl)-N-(4-(hydroxylcarbamoyl)benzyl)-1-(4-methoxyphenyl)-9H-pyrido[3,4-b]indole-3-carboxamide (9 h) is five-fold lower than that of suberoylanilide hydroxamic acid (SAHA, vorinostat). Furthermore, 9 h was found to increase the acetylation of histone H3 and α-tubulin, and to induce DNA damage as evidenced by hypochromism and enhanced phosphorylation of histone H2AX. Compound 9 h inhibits Stat3, Akt, and ERK signaling, important cell-growth-promoting pathways that are aberrantly activated in most cancers. Finally, 9 h showed reasonable solubility and permeability in Caco-2 cells. Our findings suggest that these novel β-carboline-based HDAC inhibitors may hold great promise as therapeutic agents for the treatment of human cancers.
Collapse
Affiliation(s)
- Yong Ling
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Targets, Nantong University, Nantong, 226001, P.R. China.,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, P.R. China
| | - Jiao Feng
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Targets, Nantong University, Nantong, 226001, P.R. China
| | - Lin Luo
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Targets, Nantong University, Nantong, 226001, P.R. China
| | - Jing Guo
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Targets, Nantong University, Nantong, 226001, P.R. China
| | - Yanfu Peng
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Targets, Nantong University, Nantong, 226001, P.R. China
| | - Tingting Wang
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Targets, Nantong University, Nantong, 226001, P.R. China
| | - Xiang Ge
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Targets, Nantong University, Nantong, 226001, P.R. China
| | - Qibing Xu
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Targets, Nantong University, Nantong, 226001, P.R. China
| | - Xinyang Wang
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Targets, Nantong University, Nantong, 226001, P.R. China.,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, P.R. China
| | - Hong Dai
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Targets, Nantong University, Nantong, 226001, P.R. China.,College of Chemistry and Chemical Engineering, Nantong University, Nantong, 226001, P.R. China
| | - Yanan Zhang
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Targets, Nantong University, Nantong, 226001, P.R. China
| |
Collapse
|
5
|
Synthesis and antitumor activity of novel diaryl ether hydroxamic acids derivatives as potential HDAC inhibitors. Arch Pharm Res 2012; 35:1723-32. [DOI: 10.1007/s12272-012-1003-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Revised: 03/23/2012] [Accepted: 05/14/2012] [Indexed: 12/27/2022]
|
6
|
Pang D, Zhao Y, Xue W, Shan M, Chen Y, Zhang Y, Zhang G, Liu F, Li D, Yang Y. Methylation profiles of the BRCA1 promoter in hereditary and sporadic breast cancer among Han Chinese. Med Oncol 2011; 29:1561-8. [PMID: 22076508 DOI: 10.1007/s12032-011-0100-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2011] [Accepted: 10/25/2011] [Indexed: 01/18/2023]
Abstract
The development of breast cancer is a multistep process associated with complex changes in host gene expression patterns including inactivation of tumor suppressor genes and activation of oncogenes. Critically, hereditary predisposition plays a significant role in cancer susceptibility. However, mutation of the BRCA1 gene is found only in the minority of hereditary breast cancer, which indicates that there might be alternative, novel mechanisms contributing to inactivation of the BRCA1 gene. Studies have shown that aberrant methylation of genomic DNA plays an important role in carcinogenesis. The aim of this study was to investigate whether DNA methylation may be an alternative mechanism for the inactivation of BRCA1 as an epigenetic modification of the genome and whether hereditary breast cancer has a different BRCA1 methylation phenotype pattern than sporadic breast cancer. The pattern of CpG island methylation within the promoter region of BRCA1 was assessed by bisulfite sequencing DNA from peripheral blood cells of 72 patients with hereditary predisposition but without BRCA1 mutations and 30 sporadic breast cancer controls. The overall methylation level in patients with hereditary predisposition was significantly lower than that in the sporadic control group. However, patients with hereditary predisposition showed a significantly higher methylation susceptibility for the sites -518 when compared to controls. These results suggest that there might be different BRCA1 promoter methylation levels and patterns in sporadic and hereditary breast cancer in peripheral blood DNA. These findings may facilitate the early diagnosis of hereditary breast cancer.
Collapse
Affiliation(s)
- Da Pang
- Department of Breast Surgery, The Third Affiliated Hospital of Harbin Medical University, HaPing Road 158, Harbin, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Carnero A, LLeonart ME. Epigenetic mechanisms in senescence, immortalisation and cancer. Biol Rev Camb Philos Soc 2010; 86:443-55. [DOI: 10.1111/j.1469-185x.2010.00154.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
8
|
Design and synthesis of novel isoxazole-based HDAC inhibitors. Eur J Med Chem 2010; 45:4331-8. [DOI: 10.1016/j.ejmech.2010.06.035] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2010] [Revised: 06/11/2010] [Accepted: 06/25/2010] [Indexed: 11/23/2022]
|
9
|
Histone deacetylase inhibitor induces DNA damage, which normal but not transformed cells can repair. Proc Natl Acad Sci U S A 2010; 107:14639-44. [PMID: 20679231 DOI: 10.1073/pnas.1008522107] [Citation(s) in RCA: 275] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Histone deacetylase inhibitors (HDACi) developed as anti-cancer agents have a high degree of selectivity for killing cancer cells. HDACi induce acetylation of histones and nonhistone proteins, which affect gene expression, cell cycle progression, cell migration, and cell death. The mechanism of the tumor selective action of HDACi is unclear. Here, we show that the HDACi, vorinostat (Suberoylanilide hydroxamic acid, SAHA), induces DNA double-strand breaks (DSBs) in normal (HFS) and cancer (LNCaP, A549) cells. Normal cells in contrast to cancer cells repair the DSBs despite continued culture with vorinostat. In transformed cells, phosphorylated H2AX (gammaH2AX), a marker of DNA DSBs, levels increased with continued culture with vorinostat, whereas in normal cells, this marker decreased with time. Vorinostat induced the accumulation of acetylated histones within 30 min, which could alter chromatin structure-exposing DNA to damage. After a 24-h culture of cells with vorinostat, and reculture without the HDACi, gammaH2AX was undetectable by 2 h in normal cells, while persisting in transformed cells for the duration of culture. Further, we found that vorinostat suppressed DNA DSB repair proteins, e.g., RAD50, MRE11, in cancer but not normal cells. Thus, the HDACi, vorinostat, induces DNA damage which normal but not cancer cells can repair. This DNA damage is associated with cancer cell death. These findings can explain, in part, the selectivity of vorinostat in causing cancer cell death at concentrations that cause little or no normal cell death.
Collapse
|
10
|
Ye Y, Stivers JT. Fluorescence-based high-throughput assay for human DNA (cytosine-5)-methyltransferase 1. Anal Biochem 2010; 401:168-72. [PMID: 20197058 DOI: 10.1016/j.ab.2010.02.032] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2010] [Revised: 02/23/2010] [Accepted: 02/25/2010] [Indexed: 11/30/2022]
Abstract
We have developed the first economical and rapid nonradioactive assay method that is suitable for high-throughput screening of the important pharmacological target human DNA (cytosine-5)-methyltransferase 1 (DNMT1). The method combines three key innovations: the use of a truncated form of the enzyme that is highly active on a 26-bp hemimethylated DNA duplex substrate, the introduction of the methylation site into the recognition sequence of a restriction endonuclease, and the use of a fluorogenic read-out method. The extent of DNMT1 methylation is reflected in the protection of the DNA substrate from endonuclease cleavage that would otherwise result in a large fluorescence increase. The assay has been validated in a high-throughput format, and trivial changes in the substrate sequence and endonuclease allow adaptation of the method to any bacterial or human DNA methyltransferase.
Collapse
Affiliation(s)
- Yu Ye
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | |
Collapse
|
11
|
Boulikas T. Clinical overview on Lipoplatin: a successful liposomal formulation of cisplatin. Expert Opin Investig Drugs 2010; 18:1197-218. [PMID: 19604121 DOI: 10.1517/13543780903114168] [Citation(s) in RCA: 186] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Nanoparticle formulations for packaging existing drugs have been used to treat cancer. Lipoplatin is a liposomal cisplatin encapsulated into liposome nanoparticles of an average diameter of 110 nm. Lipoplatin has substantially reduced the renal toxicity, peripheral neuropathy, ototoxicity, myelotoxicity as well as nausea/vomiting and asthenia of cisplatin in Phase I, II and III clinical studies with enhanced or similar efficacy to cisplatin. During clinical development, 10- to 200-fold higher accumulation of Lipoplatin in solid tumors compared to adjacent normal tissue was found in patients. Targeting of tumor vasculature by Lipoplatin in animals suggested its antiangiogenesis potential and Lipoplatin was proposed to act like a double-sword: as chemotherapy and an antiangiogenesis drug. Lipoplatin has finished successfully one Phase III non-inferiority clinical study as first-line against NSCLC in its combination with paclitaxel showing statistically significant reduction in nephrotoxicity; two more Phase III studies are in progress, one in NSCLC with gemcitabine also showing noninferiority with reduced toxicity and another in squamous cell carcinoma of the head and neck with 5-fluorouracil. A registrational Phase II/III study against pancreatic cancer is in progress under the orphan drug status granted to Lipoplatin by the European Medicines Agency. Phase II studies are continuing in advanced breast cancer with vinorelbine and gastrointestinal cancers with radiotherapy and 5-fluorouracil. The highlights of the clinical development of Lipoplatin are reviewed.
Collapse
Affiliation(s)
- Teni Boulikas
- Regulon, Inc., 715 N. Shoreline Blvd., Mountain View, CA 94043, USA.
| |
Collapse
|
12
|
Tapadar S, He R, Luchini DN, Billadeau DD, Kozikowski AP. Isoxazole moiety in the linker region of HDAC inhibitors adjacent to the Zn-chelating group: effects on HDAC biology and antiproliferative activity. Bioorg Med Chem Lett 2009; 19:3023-6. [PMID: 19419863 DOI: 10.1016/j.bmcl.2009.04.058] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2009] [Revised: 04/04/2009] [Accepted: 04/07/2009] [Indexed: 11/26/2022]
Abstract
A series of hydroxamic acid based histone deacetylase inhibitors 6-15, containing an isoxazole moiety adjacent to the Zn-chelating hydroxamic acid, is reported herein. Some of these compounds showed nanomolar activity in the HDAC isoform inhibitory assay and exhibited micro molar inhibitory activity against five pancreatic cancer cell lines.
Collapse
Affiliation(s)
- Subhasish Tapadar
- Department of Medicinal Chemistry & Pharmacognosy, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | | | | | | | |
Collapse
|