Novel Strategies to Mitigate Cancer Therapy Side Effects

A special issue of Cancers (ISSN 2072-6694). This special issue belongs to the section "Cancer Therapy".

Deadline for manuscript submissions: closed (22 November 2022) | Viewed by 23877

Special Issue Editor


E-Mail Website
Guest Editor
Division of Outcomes and Translational Sciences, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
Interests: clinical pharmacology; transporter; oncology; chemotherapy-induced toxicity; pharmacokinetics
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Although the effectiveness of cancer therapy has improved dramatically in the last few decades, side effects associated with small-molecule cancer drugs remain a tremendous health problem worldwide, as they negatively impact treatment outcome and diminish overall quality of life. The improved understanding of the molecular and/or cellular mechanisms that underlies the development of cancer therapy-induced side effects has resulted in continuing efforts to predict, prevent, and/or treat potentially debilitating adverse events.

In this Special Issue, we invite experts from the field to highlight novel strategies to mitigate cancer therapy-induced side effects, including both predictive and preventative interventions (including those involving genotyping, biomarker phenotyping, population PK, targeted drug treatment), as well as innovative treatment approaches (including symptom management and mechanism-based reversal of injury).

Dr. Shuiying Hu
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Cancers is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2900 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • cancer
  • oncology
  • toxicity
  • side effects
  • chemotherapy
  • small-molecule cancer drugs

Published Papers (9 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review, Other

12 pages, 3478 KiB  
Article
Ginkgolide B Regulates CDDP Chemoresistance in Oral Cancer via the Platelet-Activating Factor Receptor Pathway
by Kohei Kawasaki, Atsushi Kasamatsu, Toshiaki Ando, Tomoaki Saito, Takafumi Nobuchi, Ryunosuke Nozaki, Manabu Iyoda and Katsuhiro Uzawa
Cancers 2021, 13(24), 6299; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13246299 - 15 Dec 2021
Cited by 4 | Viewed by 2050
Abstract
The platelet-activating factor receptor (PAFR) is a key molecule that participates in intracellular signaling pathways, including regulating the activation of kinases. It is involved in cancer progression, but the detailed mechanism of its chemosensitivity is unknown. The purpose of the current study was [...] Read more.
The platelet-activating factor receptor (PAFR) is a key molecule that participates in intracellular signaling pathways, including regulating the activation of kinases. It is involved in cancer progression, but the detailed mechanism of its chemosensitivity is unknown. The purpose of the current study was to elucidate the mechanism regulating cisplatin (CDDP) sensitivity through PAFR functions in oral squamous cell carcinoma (OSCC). We first analyzed the correlation between PAFR expression and CDDP sensitivity in seven OSCC-derived cell lines based upon cell viability assays. Among them, we isolated 2 CDDP-resistant cell lines (Ca9-22 and Ho-1-N-1). In addition to conducting PAFR-knockdown (siPAFR) experiments, we found that ginkgolide B (GB), a specific inhibitor of PAFR, enhanced both CDDP chemosusceptibility and apoptosis. We next evaluated the downstream signaling pathway of PAFR in siPAFR-treated cells and GB-treated cells after CDDP treatment. In both cases, we observed decreased phosphorylation of ERK and Akt and increased expression of cleaved caspase-3. These results suggest that PAFR is a therapeutic target for modulating CDDP sensitivity in OSCC cells. Thus, GB may be a novel drug that could enhance combination chemotherapy with CDDP for OSCC patients. Full article
(This article belongs to the Special Issue Novel Strategies to Mitigate Cancer Therapy Side Effects)
Show Figures

Figure 1

13 pages, 964 KiB  
Article
Carboplatin Dosing in Children Using Estimated Glomerular Filtration Rate: Equation Matters
by Mirjam E. van de Velde, Emil den Bakker, Hester N. Blufpand, Gertjan L. Kaspers, Floor C. H. Abbink, Arjenne W. A. Kors, Abraham J. Wilhelm, Richard J. Honeywell, Godefridus J. Peters, Birgit Stoffel-Wagner, Laurien M. Buffart and Arend Bökenkamp
Cancers 2021, 13(23), 5963; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13235963 - 26 Nov 2021
Cited by 2 | Viewed by 1943
Abstract
Renal function-based carboplatin dosing using measured glomerular filtration rate (GFR) results in more consistent drug exposure than anthropometric dosing. We aimed to validate the Newell dosing equation using estimated GFR (eGFR) and study which equation most accurately predicts carboplatin clearance in children with [...] Read more.
Renal function-based carboplatin dosing using measured glomerular filtration rate (GFR) results in more consistent drug exposure than anthropometric dosing. We aimed to validate the Newell dosing equation using estimated GFR (eGFR) and study which equation most accurately predicts carboplatin clearance in children with retinoblastoma. In 13 children with retinoblastoma 38 carboplatin clearance values were obtained from individual fits using MWPharm++. Carboplatin exposure (AUC) was calculated from administered dose and observed carboplatin clearance and compared to predicted AUC calculated with a carboplatin dosing equation (Newell) using different GFR estimates. Different dosing regimens were compared in terms of accuracy, bias and precision. All patients had normal eGFR. Carboplatin exposure using cystatin C-based eGFR equations tended to be more accurate compared to creatinine-based eGFR (30% accuracy 76.3–89.5% versus 76.3–78.9%, respectively), which led to significant overexposure, especially in younger (aged ≤ 2 years) children. Of all equations, the Schwartz cystatin C-based equation had the highest accuracy and lowest bias. Although anthropometric dosing performed comparably to many of the eGFR equations overall, we observed a weight-dependent change in bias leading to underdosing in the smallest patients. Using cystatin C-based eGFR equations for carboplatin dosing in children leads to more accurate carboplatin-exposure in patients with normal renal function compared to anthropometric dosing. In children with impaired kidney function, this trend might be more pronounced. Anthropometric dosing is hampered by a weight-dependent bias. Full article
(This article belongs to the Special Issue Novel Strategies to Mitigate Cancer Therapy Side Effects)
Show Figures

Figure 1

20 pages, 50621 KiB  
Article
Memantine Protects against Paclitaxel-Induced Cognitive Impairment through Modulation of Neurogenesis and Inflammation in Mice
by Pi-Shan Sung, Pei-Wen Chen, Chia-Jui Yen, Meng-Ru Shen, Chih-Hung Chen, Kuen-Jer Tsai and Chou-Ching K. Lin
Cancers 2021, 13(16), 4177; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13164177 - 19 Aug 2021
Cited by 9 | Viewed by 2331
Abstract
Chemotherapy-induced cognitive impairment (CICI) is an adverse side effect of cancer treatment with increasing awareness. Hippocampal damage and related neurocognitive impairment may mediate the development of CICI, in which altered neurogenesis may play a role. In addition, increased inflammation may be related to [...] Read more.
Chemotherapy-induced cognitive impairment (CICI) is an adverse side effect of cancer treatment with increasing awareness. Hippocampal damage and related neurocognitive impairment may mediate the development of CICI, in which altered neurogenesis may play a role. In addition, increased inflammation may be related to chemotherapy-induced hippocampal damage. Memantine, an uncompetitive N-methyl-D-aspartate (NMDA) receptor antagonist that may enhance neurogenesis and modulate inflammation, may be useful for treating CICI. To test this hypothesis, paclitaxel was administered to eight-week-old male B6 mice to demonstrate the relationship between CICI and impaired neurogenesis, and then, we evaluated the impact of different memantine regimens on neurogenesis and inflammation in this CICI model. The results demonstrated that both the pretreatment and cotreatment regimens with memantine successfully reversed impaired neurogenesis and spatial memory impairment in behavior tests. The pretreatment regimen unsuccessfully inhibited the expression of peripheral and central TNF-α and IL-1β and did not improve the mood alterations following paclitaxel treatment. However, the cotreatment regimen led to a better modulatory effect on inflammation and restoration of mood disturbance. In conclusion, this study illustrated that impaired neurogenesis is one of the mechanisms of paclitaxel-induced CICI. Memantine may serve as a potential treatment for paclitaxel-induced CICI, but different treatment strategies may lead to variations in the treatment efficacy. Full article
(This article belongs to the Special Issue Novel Strategies to Mitigate Cancer Therapy Side Effects)
Show Figures

Figure 1

Review

Jump to: Research, Other

22 pages, 2516 KiB  
Review
Application Prospects of Triphenylphosphine-Based Mitochondria-Targeted Cancer Therapy
by Xiaoxia Cheng, Dong Feng, Junyu Lv, Xiaoman Cui, Yichen Wang, Qun Wang and Lei Zhang
Cancers 2023, 15(3), 666; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers15030666 - 21 Jan 2023
Cited by 18 | Viewed by 3231
Abstract
Cancer is one of the leading causes of death and the most important impediments to the efforts to increase life expectancy worldwide. Currently, chemotherapy is the main treatment for cancer, but it is often accompanied by side effects that affect normal tissues and [...] Read more.
Cancer is one of the leading causes of death and the most important impediments to the efforts to increase life expectancy worldwide. Currently, chemotherapy is the main treatment for cancer, but it is often accompanied by side effects that affect normal tissues and organs. The search for new alternatives to chemotherapy has been a hot research topic in the field of antineoplastic medicine. Drugs targeting diseased tissues or cells can significantly improve the efficacy of drugs. Therefore, organelle-targeted antitumor drugs are being explored, such as mitochondria-targeted antitumor drugs. Mitochondria is the central site of cellular energy production and plays an important role in cell survival and death. Moreover, a large number of studies have shown a close association between mitochondrial metabolism and tumorigenesis and progression, making mitochondria a promising new target for cancer therapy. Combining mitochondrial targeting agents with drug molecules is an effective way of mitochondrial targeting. In addition, hyperpolarized tumor cell membranes and mitochondrial membrane potentially allow selective accumulation of mitochondria-targeted drugs. This enhances the direct killing of tumor cells by drug molecules while minimizing the potential toxicity to normal cells. In this review, we discuss the common pro-mitochondrial agents, the advantages of triphenylphosphine (TPP) in mitochondrial-targeted cancer therapy and systematically summarize various TPP-based mitochondria-targeting anticancer drugs. Full article
(This article belongs to the Special Issue Novel Strategies to Mitigate Cancer Therapy Side Effects)
Show Figures

Figure 1

15 pages, 665 KiB  
Review
Pharmacogenomics for Prediction of Cardiovascular Toxicity: Landscape of Emerging Data in Breast Cancer Therapies
by Renske Altena, Svetlana Bajalica-Lagercrantz and Andri Papakonstantinou
Cancers 2022, 14(19), 4665; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers14194665 - 25 Sep 2022
Cited by 3 | Viewed by 1876
Abstract
Pharmacogenomics is an emerging field in oncology, one that could provide valuable input on identifying patients with inherent risk of toxicity, thus allowing for treatment tailoring and personalization on the basis of the clinical and genetic characteristics of a patient. Cardiotoxicity is a [...] Read more.
Pharmacogenomics is an emerging field in oncology, one that could provide valuable input on identifying patients with inherent risk of toxicity, thus allowing for treatment tailoring and personalization on the basis of the clinical and genetic characteristics of a patient. Cardiotoxicity is a well-known side effect of anthracyclines and anti-HER2 agents, although at a much lower incidence for the latter. Data on single-nucleotide polymorphisms related to cardiotoxicity are emerging but are still scarce, mostly being of retrospective character and heterogeneous. A literature review was performed, aiming to describe current knowledge in pharmacogenomics and prediction of cardiotoxicity related to breast cancer systemic therapies and radiotherapies. Most available data regard genes encoding various enzymes related to anthracycline metabolism and HER2 polymorphisms. The available data are presented, together with the challenges and open questions in the field. Full article
(This article belongs to the Special Issue Novel Strategies to Mitigate Cancer Therapy Side Effects)
Show Figures

Figure 1

15 pages, 294 KiB  
Review
Bridging the Translational Gap in Chemotherapy-Induced Peripheral Neuropathy with iPSC-Based Modeling
by Christina Mortensen, Nanna Elman Andersen and Tore Bjerregaard Stage
Cancers 2022, 14(16), 3939; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers14163939 - 15 Aug 2022
Cited by 4 | Viewed by 1698
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a common and potentially serious adverse effect of a wide range of chemotherapeutics. The lack of understanding of the molecular mechanisms underlying CIPN limits the efficacy of chemotherapy and development of therapeutics for treatment and prevention of CIPN. [...] Read more.
Chemotherapy-induced peripheral neuropathy (CIPN) is a common and potentially serious adverse effect of a wide range of chemotherapeutics. The lack of understanding of the molecular mechanisms underlying CIPN limits the efficacy of chemotherapy and development of therapeutics for treatment and prevention of CIPN. Human induced pluripotent stem cells (iPSCs) have become an important tool to generate the cell types associated with CIPN symptoms in cancer patients. We reviewed the literature for iPSC-derived models that assessed neurotoxicity among chemotherapeutics associated with CIPN. Furthermore, we discuss the gaps in our current knowledge and provide guidance for selecting clinically relevant concentrations of chemotherapy for in vitro studies. Studies in iPSC-derived neurons revealed differential sensitivity towards mechanistically diverse chemotherapeutics associated with CIPN. Additionally, the sensitivity to chemotherapy was determined by donor background and whether the neurons had a central or peripheral nervous system identity. We propose to utilize clinically relevant concentrations that reflect the free, unbound fraction of chemotherapeutics in plasma in future studies. In conclusion, iPSC-derived sensory neurons are a valuable model to assess CIPN; however, studies in Schwann cells and motor neurons are warranted. The inclusion of multiple iPSC donors and concentrations of chemotherapy known to be achievable in patients can potentially improve translational success. Full article
(This article belongs to the Special Issue Novel Strategies to Mitigate Cancer Therapy Side Effects)
13 pages, 442 KiB  
Review
Testing for Dihydropyrimidine Dehydrogenase Deficiency to Individualize 5-Fluorouracil Therapy
by Robert B. Diasio and Steven M. Offer
Cancers 2022, 14(13), 3207; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers14133207 - 30 Jun 2022
Cited by 13 | Viewed by 4717
Abstract
Severe adverse events (toxicity) related to the use of the commonly used chemotherapeutic drug 5-fluorouracil (5-FU) affect one in three patients and are the primary reason cited for premature discontinuation of therapy. Deficiency of the 5-FU catabolic enzyme dihydropyrimidine dehydrogenase (DPD, encoded by [...] Read more.
Severe adverse events (toxicity) related to the use of the commonly used chemotherapeutic drug 5-fluorouracil (5-FU) affect one in three patients and are the primary reason cited for premature discontinuation of therapy. Deficiency of the 5-FU catabolic enzyme dihydropyrimidine dehydrogenase (DPD, encoded by DPYD) has been recognized for the past 3 decades as a pharmacogenetic syndrome associated with high risk of 5-FU toxicity. An appreciable fraction of patients with DPD deficiency that receive 5-FU-based chemotherapy die as a result of toxicity. In this manuscript, we review recent progress in identifying actionable markers of DPD deficiency and the current status of integrating those markers into the clinical decision-making process. The limitations of currently available tests, as well as the regulatory status of pre-therapeutic DPYD testing, are also discussed. Full article
(This article belongs to the Special Issue Novel Strategies to Mitigate Cancer Therapy Side Effects)
Show Figures

Figure 1

29 pages, 1157 KiB  
Review
Solute Carrier Nucleoside Transporters in Hematopoiesis and Hematological Drug Toxicities: A Perspective
by Syed Saqib Ali, Ruchika Raj, Tejinder Kaur, Brenna Weadick, Debasis Nayak, Minnsung No, Jane Protos, Hannah Odom, Kajal Desai, Avinash K. Persaud, Joanne Wang and Rajgopal Govindarajan
Cancers 2022, 14(13), 3113; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers14133113 - 25 Jun 2022
Cited by 3 | Viewed by 2178
Abstract
Anticancer nucleoside analogs produce adverse, and at times, dose-limiting hematological toxicities that can compromise treatment efficacy, yet the mechanisms of such toxicities are poorly understood. Recently, cellular nucleoside transport has been implicated in normal blood cell formation with studies from nucleoside transporter-deficient mice [...] Read more.
Anticancer nucleoside analogs produce adverse, and at times, dose-limiting hematological toxicities that can compromise treatment efficacy, yet the mechanisms of such toxicities are poorly understood. Recently, cellular nucleoside transport has been implicated in normal blood cell formation with studies from nucleoside transporter-deficient mice providing additional insights into the regulation of mammalian hematopoiesis. Furthermore, several idiopathic human genetic disorders have revealed nucleoside transport as an important component of mammalian hematopoiesis because mutations in individual nucleoside transporter genes are linked to various hematological abnormalities, including anemia. Here, we review recent developments in nucleoside transporters, including their transport characteristics, their role in the regulation of hematopoiesis, and their potential involvement in the occurrence of adverse hematological side effects due to nucleoside drug treatment. Furthermore, we discuss the putative mechanisms by which aberrant nucleoside transport may contribute to hematological abnormalities and identify the knowledge gaps where future research may positively impact treatment outcomes for patients undergoing various nucleoside analog therapies. Full article
(This article belongs to the Special Issue Novel Strategies to Mitigate Cancer Therapy Side Effects)
Show Figures

Figure 1

Other

Jump to: Research, Review

21 pages, 1539 KiB  
Perspective
Targeting Histone Epigenetic Modifications and DNA Damage Responses in Synthetic Lethality Strategies in Cancer?
by Pedro A. Lazo
Cancers 2022, 14(16), 4050; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers14164050 - 22 Aug 2022
Cited by 8 | Viewed by 2662
Abstract
Synthetic lethality strategies are likely to be integrated in effective and specific cancer treatments. These strategies combine different specific targets, either in similar or cooperating pathways. Chromatin remodeling underlies, directly or indirectly, all processes of tumor biology. In this context, the combined targeting [...] Read more.
Synthetic lethality strategies are likely to be integrated in effective and specific cancer treatments. These strategies combine different specific targets, either in similar or cooperating pathways. Chromatin remodeling underlies, directly or indirectly, all processes of tumor biology. In this context, the combined targeting of proteins associated with different aspects of chromatin remodeling can be exploited to find new alternative targets or to improve treatment for specific individual tumors or patients. There are two major types of proteins, epigenetic modifiers of histones and nuclear or chromatin kinases, all of which are druggable targets. Among epigenetic enzymes, there are four major families: histones acetylases, deacetylases, methylases and demethylases. All these enzymes are druggable. Among chromatin kinases are those associated with DNA damage responses, such as Aurora A/B, Haspin, ATM, ATR, DNA-PK and VRK1—a nucleosomal histone kinase. All these proteins converge on the dynamic regulation chromatin organization, and its functions condition the tumor cell viability. Therefore, the combined targeting of these epigenetic enzymes, in synthetic lethality strategies, can sensitize tumor cells to toxic DNA-damage-based treatments, reducing their toxicity and the selective pressure for tumor resistance and increasing their immunogenicity, which will lead to an improvement in disease-free survival and quality of life. Full article
(This article belongs to the Special Issue Novel Strategies to Mitigate Cancer Therapy Side Effects)
Show Figures

Figure 1

Back to TopTop