Skip to main content
Genes & Diseases logoLink to Genes & Diseases
. 2022 Mar 18;10(4):1367–1401. doi: 10.1016/j.gendis.2022.02.007

Cancer chemotherapy and beyond: Current status, drug candidates, associated risks and progress in targeted therapeutics

Uttpal Anand a, Abhijit Dey b,1, Arvind K Singh Chandel c,1, Rupa Sanyal d, Amarnath Mishra e,∗∗, Devendra Kumar Pandey f, Valentina De Falco g, Arun Upadhyay h, Ramesh Kandimalla i,j, Anupama Chaudhary k, Jaspreet Kaur Dhanjal l, Saikat Dewanjee m, Jayalakshmi Vallamkondu n, José M Pérez de la Lastra o,
PMCID: PMC10310991  PMID: 37397557

Abstract

Cancer is an abnormal state of cells where they undergo uncontrolled proliferation and produce aggressive malignancies that causes millions of deaths every year. With the new understanding of the molecular mechanism(s) of disease progression, our knowledge about the disease is snowballing, leading to the evolution of many new therapeutic regimes and their successive trials. In the past few decades, various combinations of therapies have been proposed and are presently employed in the treatment of diverse cancers. Targeted drug therapy, immunotherapy, and personalized medicines are now largely being employed, which were not common a few years back. The field of cancer discoveries and therapeutics are evolving fast as cancer type-specific biomarkers are progressively being identified and several types of cancers are nowadays undergoing systematic therapies, extending patients' disease-free survival thereafter. Although growing evidence shows that a systematic and targeted approach could be the future of cancer medicine, chemotherapy remains a largely opted therapeutic option despite its known side effects on the patient's physical and psychological health. Chemotherapeutic agents/pharmaceuticals served a great purpose over the past few decades and have remained the frontline choice for advanced-stage malignancies where surgery and/or radiation therapy cannot be prescribed due to specific reasons. The present report succinctly reviews the existing and contemporary advancements in chemotherapy and assesses the status of the enrolled drugs/pharmaceuticals; it also comprehensively discusses the emerging role of specific/targeted therapeutic strategies that are presently being employed to achieve better clinical success/survival rate in cancer patients.

Keywords: Antimicrobial peptides, Cancer therapies, Clinical trials, Combination therapy, Immunotherapy, Patient survival, Personalized medicine, Targeted drug delivery

Introduction

Cancer is a heterogeneous and multifactorial disease in which a series of genomic/molecular alterations cause the uncontrolled growth and proliferation of the cells, causing a rapid increase in tissue mass in the affected parts of the body. Under normal conditions, a cell gets signals to die and to supplant the organism with a young and healthier cell. Cancer cells grow using the body's oxygen and supplements, depriving other cells of regular supplements and growth factors. These cells can turn the microenvironment in their favor, deceive the immune system of the body, and can exploit the physiology of other cells to accommodate their needs.1, 2, 3, 4, 5, 6 Some of the biomarkers which are currently been used to detect cancer including human epididymis protein 4 (HE4), carcinoembryonic antigen (CEA), legumain, mesothelin, osteopontin, and vitamin E-binding plasma protein. A plethora of anti-cancer drugs and natural medicinal compounds have been devised over the years, which can suppress tumor growth through diverse mechanisms. Some of the drugs/compounds work on crucial cellular enzymes, while others may alter cell metabolism. They have also shown the potential to interfere with some critical cellular processes, e.g., programmed cell death/apoptosis, drug resistance, DNA damage, DNA replication, or immune reactions. These drugs have distinct modes of action and selectivity for multiple cancer types7, 8, 9, 10, 11, 12; however, subtle alteration to their chemical structure could abolish their anti-cancer potency.13,14

The idea of chemotherapy (utilizing toxic compounds and drugs to destroy cancerous cells) came into existence after the reports of mustard gas killing lymphatic tissues and bone marrow. The effects were later validated in mice using an effective derivative of the gas (nitrogen mustard) that showed effective regression of lymphoma tissues.15, 16, 17 The first patient receiving this nitrogen mustard was forty-eight years old lymphosarcoma patient whose cancer softened and cleared initially. Unfortunately, later he died after relapsing, but this secret military trial at Yale University paved the way for chemicals in treating cancers and developing the field of cancer chemotherapy for treating a variety of cancers.18, 19, 20, 21

Chemotherapy primarily works by circumventing the cancer cells from further growth and division. Cancer cells usually divide and grow at a much-accelerated rate than normal cells and are physiologically possess very high endogenous stress. Therefore, the drugs can destroy them rapidly and more effectively in comparison to other surrounding cells. Some of the promising inhibitor therapies which have been used to treat solid cancers are polyadenosine diphosphate-ribose polymerase inhibitors, angiogenesis inhibitors, histone deacetylase (HDAC) inhibitors, mechanistic target of rapamycin (mTOR) inhibitors, poly (adenosine di-phosphate-ribose) polymerase (PARP) inhibitors, p53/mouse double minute 2 homolog (MDM2) inhibitors, hedgehog pathway blockers, tyrosine kinase inhibitors and proteasome inhibitors.22,23 Chemotherapy may have different variations, which can affect the target cells in distinct manners.24 Some of the treatments may directly alter the quality of cellular proteins, rendering them non-functional and affecting major cellular physiological pathways. Major chaperone repressors, autophagy suppressors, or proteasome inhibitors belong to these classes of molecules.25, 26, 27, 28, 29, 30, 31 Other drugs may target some essential hormones and interfere with the body's overall metabolism.32,33 The 2018 Nobel Prize attributed to immune checkpoint therapy research has highlighted the importance of immunotherapy, which underlies the possibilities of modulating our immune system in our favor to cope up with the cancer-like conditions and fight back against the disease.24,34

The selection of chemo-preventive drugs or a combination of drugs is mostly based on the type and stage of cancer. The primary objective of these drugs is to neutralize the cancerous cells and ameliorate the stress caused by the tumor's growth. The dosage and timespan of the treatment are two important points of consideration. It has been observed that the medications are given at very high doses causing numerous side effects and harm to other healthy cells.35 One major challenge is the relapse of the disease. During reoccurrence, it has been widely observed that the cancer cells attain drug resistance following longer exposure to the drugs. Despite enormous applications, most chemotherapies are given to prolong and ease the patient's survival, hence called palliative chemotherapy.21 However, prolonged exposure to these drugs may have excessively adverse effects on a patient's physical and mental health, making it difficult to continue the ongoing treatment. Oncologists, for the most part, provide these medications with possible time intervals in between, which also gives non-transformed cells a chance to mend.36 In most cases, a team of specialists, including radiologists, nutritionists, psychologists, and the primary consultant collectively decide the treatment plans that include drug combinations, dosage, duration of cycles, and additional supplements.

This report mainly reviews various aspects of cancer chemotherapy, including various applications of the drugs/chemotherapeutic agents that are already in practice. We also succinctly discussed various side effects and drawbacks of these treatment plans and further provided an overview of recent advancements in interventional strategies by focusing on targeted drug therapy, immunotherapy, personalized and integrative medicines. Figure 1 depicts the significant intrinsic and extrinsic cellular and molecular events causing the transformation of a normal cell to a cancer cell and its distinct hallmarks.

Figure 1.

Fig. 1

Schematic diagram showing different intrinsic and extrinsic biological/molecular events potentiating the transformation of a normal cell to the cancer cell, while the lower part of this diagram showing different hallmarks of a transformed cancer cell.

Cancer chemotherapeutics: what is known so far?

The idea of cancer as a disease was known to Hippocrates, the father of medicine, around 400 BC. He later coined the term ‘carcinos’ due to the similarity of cancer tissue growths with crabs. The term was later replaced with the Latin word cancer by Celsus. Many ancient pieces of literature have described several kinds of malignancies in humans, and multiple treatment approaches were also in use in different parts of the ancient world, as found in the literature of that era.37,38 However, the progress of strategies to treat the disease was more or less static until the beginning of the twentieth century.39 The extract of the plant Colchicum autumnale was used by Greek physicians to dissolve the tumor mass. Interestingly, in the 1930s, the active compound in the extract, colchicine, was reported that can interfere with microtubule assembly and may work as a potential drug.40 Vincristine and vinblastine are other similar compounds that were medicinally recognized as possible anti-tumor agents a long time ago.41 The ancient pieces of the literature suggest that the Greek and Roman physicians have contributed enormously to the identification and characterization of disease. Arabic, Indian, and Chinese medicinal archives have also paved the way for multiple medieval and modern approaches to understand and treat the disease. Interestingly, for centuries, most of the approaches remain limited to cauterization, bloodletting, surgery, herbal medicine, etc.42 In modern onco-medicine, which was evolved during the Second World War, many active compounds were reported following the detailed characterization of the disease at the genetic and molecular level. The initial decades have seen enormous debate on the use of cytotoxic agents as drug candidates; however, advancements were made, and success came in the form of Eli Lily introducing Vinca alkaloids in the 1960s, which was then followed by procarbazine.43 In the following years, the idea of combination therapy was introduced for acute lymphocytic leukemia and non-Hodgkin's lymphoma patients that were also supplemented with supportive medical care to enhance the quality of life.21

In the second half of the twentieth century, a series of reports have identified numerous mutations, external stimuli (toxic chemicals, viruses, etc.), metabolic alterations, etc. as the underlying causes of cancer.44,45 Identification of a plethora of cellular pathways that are affected in various cancer types has given different molecular alternatives to target and develop effective drugs (Fig. 2).46, 47, 48 Several modes of medications have also been introduced and tried for drug administration based on the types and stages of the diagnosed cancer. Here, we discuss the available drug medications, following which we have revisited the chemotherapeutic agents, which have substantially pushed the field forward and extended the lives of millions in the past several decades.

Figure 2.

Fig. 2

Schematic diagram showing the basic principles of chemotherapy (A), different classes of chemotherapeutic agents/drugs (B), and immunotherapeutic mechanism(s) of targeting cancer cells (C).

Intravenous (IV) chemotherapy requires drug infusion straightforwardly into the patient's veins that may take from minutes to a few hours. Some medications can also be given either as pills or fluid regularly or at some intervals.49 Intramuscular shots and intra-abdominal administration are other methods of giving chemotherapeutic medications to target the specific locations of tumor mass.50 In addition, a new technology called pressurized intraperitoneal aerosol chemotherapy has been developed to efficiently deliver intraperitoneal chemotherapy for end-stage peritoneal metastases patients.51 Topical treatment methods imply providing drugs through the skin surface. The anti-cancer compounds derived from either natural sources or their synthetic chemical analogs can kill cancer cells or stop their development.52, 53, 54 Notably, various other approaches are emerging to treat the cancer cells, including small molecule inhibitors,55 a combination of anti-cancer agents and immunotherapy,56 nanotechnology-based drugs,57, 58, 59 and organo-seleno compounds (Fig. 2).31

Nevertheless, the selectivity of most drug particles is restricted, and such medications are considered one of the most harmful medications utilized in the treatment.60,61 Since the initial findings of modern anticancer drug discoveries in the 1940s, continuous efforts are being made to treat this life-threatening disease. Advances in scientific methods and techniques have brought new treatment and diagnostic tools and advanced patient care measures (Fig. 2).62, 63, 64, 65, 66, 67, 68, 69 Treatment plans may have palliation as one strategy that may lead to shrinkage of the obvious tumor, easing side effects, and increasing the life quality of patients. Figure 2 describes various ways of providing anti-cancer medications to patients depending upon their type and stage of the disease.

Along with primary treatment, adjuvant therapy is also given to the patients for additionally combating the disease through supportive strategies.70,71 Surgery and radiation can be given to patients to supplement a chemotherapeutic plan to curb the disease by destroying shrouded cancer cells that could not be targeted otherwise.72 Sometimes adjuvant chemotherapy is given to patients after surgery or radiation or both to lessen the chances of recurrence.73, 74, 75 Adjuvant chemotherapy starts typically within three to five weeks of the surgical removal of a tumor and has distinctive treatment lengths relying upon the disease. Although chemotherapy has significantly improved overall survival, patients experience a wide range of physical and psychological symptoms that impact their quality of life. Symptoms seldom occur in isolation. Chemotherapy-induced hair loss remains greatly feared, with a negative impact on the well-being of many cancer patients. Cancer- and chemotherapy-induced poor appetite is usually the result of taste changes, mouth sores, nausea and vomiting, increased satiety, medication side effects, pain, fatigue, depressed mood, and anxiety.

Basic principles of chemotherapy: years of research, miles to go

The cancer cell metabolic mechanisms functionally overlap with the host cells, so cancer treatment is very challenging.76,77 Designing drugs or therapeutics mainly focuses on selectivity, which can specifically kill the cancerous cells without affecting the non-cancerous cells (Fig. 2). In a way, the anti-cancer therapy may resemble antimicrobials, but the cancerous cells and the bacterial cells have differences in terms of the metabolic and physiological characters.78, 79, 80 For our immune cells, bacterial cell recognition is straightforward, while our transformed cells can hide cell surface receptors to evade the immune system.81 Early-stage detection of cancers increases the chances of treatment and survival. Therefore, recognizing and diagnosing those transformed cells remains one of the most challenging tasks in oncological research. At early stages, most known biomarkers remain undetectable, and cancer cells' ability to hide from immune cells freezes our immune responses.82,83

In a tumor subpopulation, transformed cells differ drastically from the other cells in terms of physiology, metabolism, proliferation rate, etc.84, 85, 86 Besides that, the genetic diversity may pose additional challenges to target all kinds of cells in a particular tissue mass owing to the tumor heterogeneity.87,88 A combination of drug therapy is applied to address this challenge, and the regime is devised according to the cancer phase and type (Fig. 2).89,90

In the subsequent section, we provide a comprehensive overview of various chemotherapeutic agents which have served the purpose for several decades in different cancer types. These drugs could be classified in different ways based on their chemical nature/source, molecular target, mechanism/mode of action, or effectiveness in model systems against various malignancies/cancer types (Table 1).

Table 1.

Table listing chemotherapeutic drugs/agents used in current clinical practice along with details of their molecular targets, mode of action, and investigated model systems across diverse malignancies.

Drugs/Compound Molecular targets Mechanism of action Studied model system Reference/Source
Antineoplastic alkylating agents
Altretamine DNA polymerase alpha catalytic subunit
Ribonucleoside-diphosphate reductase large subunit
DNA
Inhibitor
Inhibitor
Other/unknow
B-cell chronic lymphocytic, leukemia/prolymphocytic leukemia/small lymphocytic lymphoma refractory, and refractory non-Hodgkin's lymphoma 278
Bendamustine DNA Intra- and inter-strand crosslinking Chronic lymphocytic leukaemia (CLL), follicular non-Hodgkin's lymphoma refractory, refractory Hodgkin lymphoma, refractory Mantle cell lymphoma, Waldenström's macroglobulinemia (WM), recurrent multiple myeloma, and refractory indolent B cell non-Hodgkin lymphoma 279
Busulfan DNA Cross-linking/alkylation Chronic myelogenous leukemia 279
Carboplatin DNA Cross-linking/alkylation Advanced cervical cancer, advanced Endometrial Cancer, advanced esophageal cancers, advanced head and neck Cancer, advanced melanoma, advanced non-small cell lung cancer, advanced ovarian carcinoma, advanced Sarcoma, metastatic breast cancer, neuroendocrine carcinoma of the skin, pleural mesothelioma, refractory Hodgkin lymphoma, retinoblastoma, advanced bladder cancer, advanced small cell lung cancer, advanced testicular cancer, advanced thymoma, advanced thymic carcinoma, and refractory non-Hodgkin's lymphoma 279
Carmustine DNA
Glutathione reductase, mitochondrial
RNA
Other/unknown inhibitor
Other/unknown
Glioblastoma, brainstem glioma, medulloblastoma, astrocytoma, ependymoma and metastatic brain tumors, multiple myeloma, Hodgkin's disease, non-Hodgkin's lymphomas, and may be used on the skin (topically) for cutaneous T-cell lymphoma 280
Chlorambucil DNA Cross-linking/alkylation Chronic lymphocytic leukemia, lymphomas, and Hodgkin's disease 279
Cisplatin DNA
DNA-3-methyladenine glycosylase
Alpha-2-macroglobulin
Serotransferrin
Copper transport protein ATOX1
Cross-linking/alkylation
Not Available
Not Available
Not Available
Not Available
Testicular cancer, ovarian cancer, bladder cancer, head and neck cancer, esophagus cancer, small cell and non-small cell lung cancer, non-Hodgkin's lymphoma and trophoblastic neoplasms 279
Cyclophosphamide DNA
Nuclear receptor subfamily 1 group I member 2
Cross-linking/alkylation
Not Available
Lymphoma, multiple myeloma, leukemia, ovarian cancer, breast cancer, small cell lung cancer, neuroblastoma, and sarcoma. 279
Dacarbazine DNA
DNA polymerase alpha subunit B
6-phosphogluconate dehydrogenase, decarboxylating
Cross-linking/alkylation
Other/unknown
Inhibitor
Melanoma skin cancer, soft tissue sarcoma, Hodgkin lymphoma 279
Daunorubicin DNA
DNA topoisomerase 2-alpha
DNA topoisomerase 2-beta
Intercalation
Inhibitor
Inhibitor
Acute nonlymphocytic leukemia in adults and acute lymphocytic leukemia in adults and children 279
Decitabine DNA
DNA (cytosine-5)-methyltransferase 1
DNA (cytosine-5)-methyltransferase 3A
DNA (cytosine-5)-methyltransferase 3B
Other/unknown
Inhibitor
Inhibitor
Inhibitor
Myelodysplastic syndromes, and acute myeloid leukemia (AML) 279,281
Doxorubicin DNA
DNA topoisomerase 2-alpha
Nucleolar and coiled-body phosphoprotein 1
Intercalation
Inhibitor
Not available
279
Epirubicin DNA topoisomerase 2-alpha
DNA
Inhibitor
Intercalation
Breast cancer 279,282
Etoposide DNA topoisomerase 2-alpha
DNA topoisomerase 2-beta
inhibitor
Inhibitor
Testicular cancer, lung cancer, lymphoma, nonlymphocytic leukemia, neuroblastoma, and ovarian cancer, Kaposi's sarcoma, Ewing's sarcoma and glioblastoma multiforme 279
Idarubicin DNA
DNA topoisomerase 2-alpha
Intercalation
Inhibitor
Acute myelogenous, acute lymphoblastic and chronic myelogenous 279
Ifosfamide DNA
Nuclear receptor subfamily 1 group I member 2
Other/unknown
Not available
Germ cell testicular cancer, soft tissue sarcoma, acute lymphocytic leukemia, bladder cancer, bone cancer, pancreatic cancer, stomach cancer, breast cancer, cervical cancer, endometrial cancer, lung cancer, ovarian cancer, neuroblastoma, lymphoma, and Wilms' tumor 279
Irinotecan DNA topoisomerase 1,
DNA topoisomerase I, mitochondrial
Inhibitor
Inhibitor
Metastatic colorectal cancer, other colorectal cancers and non-small/small lung cancers 279
Lomustine DNA
Stathmin-4
Cross-linking/alkylation
Antagonist
Metastatic brain tumors, Hodgkin's and non-Hodgkin's lymphoma 279
Mechlorethamine DNA Intercalation Hodgkin's disease, chronic lymphocytic leukemia, chronic myelogenous leukemia, small cell lung cancer, medulloblastoma, and non-Hodgkin's lymphoma 283
Melphalan DNA Cross-linking/alkylation Multiple myeloma, ovarian cancer, breast cancer, melanoma, and amyloidosis 284
Mitomycin C DNA Cross-linking/alkylation Gastric cancer, anal and colon cancer, breast cancer, non-small cell lung cancer, head and neck cancer, small bladder papillomas, pancreatic cancer, cervical cancer 279
Mitoxantrone DNA
DNA topoisomerase 2-alpha
Intercalation
Inhibitor
Advanced, hormone-refractory prostate cancer, acute nonlymphocytic leukemia (ANLL), breast cancer and non-Hodgkin's lymphoma. 285
Oxaliplatin DNA Cross-linking/alkylation Metastatic or recurrent colorectal cancer, metastatic pancreatic cancer and metastatic gastric cancer. 286
Temozolomide DNA Cross-linking/alkylation Brain tumors (astrocytomas) in adult patients, metastatic melanoma and glioblastoma multiforme 287
Topotecan DNA topoisomerase 1
DNA topoisomerase I, mitochondrial DNA
Inhibitor
Inhibitor
Intercalation
Ovarian cancer, small cell lung cancer, and certain types of cervical cancer 288
Thiotepa DNA Cross-linking/alkylation Adenocarcinoma of the breast, adenocarcinoma of the ovary, papillary thyroid cancer and bladder cancer, Hodgkin's and non-Hodgkin's lymphomas 289
Trabectedin DNA Binder Soft tissue sarcoma, and ovarian cancer 290
Antineoplastic antimetabolites
Azathioprine Ras-related C3 botulinum toxin substrate 1 Modulator Leukemia and lymphomas 291
Cladribine Ribonucleoside-diphosphate reductase large subunit
Ribonucleoside-diphosphate reductase subunit M2
Ribonucleoside-diphosphate reductase subunit M2 B
DNA
DNA polymerase alpha catalytic subunit
DNA polymerase epsilon catalytic subunit A
DNA polymerase epsilon subunit 2
DNA polymerase epsilon subunit 3
DNA polymerase epsilon subunit 4
Purine nucleoside phosphorylase
Inhibitor
Inhibitor
Inhibitor
Other/unknown
Inhibitor
Inhibitor
Inhibitor
Inhibitor
Inhibitor
Inducer
Drug-resistant T-cell prolymphocytic leukemia 292
Clofarabine DNA polymerase alpha catalytic subunit
Ribonucleoside-diphosphate reductase large subunit
DNA
Inhibitor
Inhibitor
Other/unknown
Relapsed or refractory acute lymphoblastic leukaemia, acute myeloid leukaemia, juvenile myelomonocytic leukaemia, and myelodysplastic syndrome 293
Fludarabine Ribonucleoside-diphosphate reductase large subunit
DNA polymerase alpha catalytic subunit
DNA
Deoxycytidine kinase
Inhibitor
Inhibitor
Incorporation into and destabilization
Agonist
Chronic lymphocytic leukemia, non-Hodgkin's lymphoma, acute myeloid leukemia, and acute lymphocytic leukemia 294
Gemcitabine DNA
Ribonucleoside-diphosphate reductase large subunit
Thymidylate synthase
UMP-CMP kinase
Cross-linking/alkylation
Inhibitor
Inhibitor
Inhibitor
Breast cancer, ovarian cancer, non-small cell lung cancer, pancreatic cancer and bladder cancer 295
Mercaptopurine Hypoxanthine-guanine phosphoribosyltransferase
Amidophosphoribosyltransferase
Inosine-5′-monophosphate dehydrogenase
Inhibitor
Inhibitor
Inhibitor
Acute lymphocytic leukemia (ALL), chronic myeloid leukemia (CML), Crohn's disease and ulcerative colitis. 296
Nelarabine DNA
DNA polymerase alpha catalytic subunit
Incorporation into and destabilization
Inhibitor
Acute T-cell lymphoblastic leukemia 297
Pentostatin Adenosine deaminase inhibitor Hairy cell leukaemia refractory to alpha interferon 298
Tioguanine DNA Intercalation Acute leukemia 299
Pyrimidine-derived antimetabolites (pyrimidine antagonists)
Capecitabine Thymidylate synthase DNA
RNA
Inhibitor
Incorporation into and destabilization
Incorporation into and destabilization
Metastatic breast, gastric and colorectal cancers 99
Cytarabine DNA polymerase beta
DNA
Inhibitor
Cross-linking/alkylation
Acute myeloid leukemia (AML), acute lymphocytic leukemia (ALL), chronic myelogenous leukemia (CML) and non-Hodgkin's lymphoma 97
Decitabine DNA
DNA (cytosine-5)-methyltransferase 1
DNA (cytosine-5)-methyltransferase 3A
DNA (cytosine-5)-methyltransferase 3B
Other/unknown
Inhibitor
Inhibitor
Inhibitor
Myelodysplastic syndromes (MDS) 101
Gemcitabine DNA
Ribonucleoside-diphosphate reductase large subunit
Thymidylate synthase
UMP-CMP kinase
Cross-linking/alkylation
Inhibitor
Inhibitor
Inhibitor
Metastatic ovarian cancer, metastatic non-small cell lung cancer, and metastatic pancreatic adenocarcinoma 100
Fluorouracil Thymidylate synthase
DNA
RNA
Other/unknown
Incorporation into and destabilization
Incorporation into and destabilization
Actinic keratosis (AK), breast cancer, malignant neoplasm of colon, malignant neoplasm of pancreas, malignant neoplasm of stomach, rectal carcinoma, superficial basal cell carcinoma, and hyperkeratotic actinic keratosis 98
Raltitrexed Thymidylate synthase
Folylpolyglutamate synthase, mitochondrial
Inhibitor
Antagonist
Advanced Colorectal Cancer, Pleural Mesotheliomas 102
Tegafur Thymidylate synthase Inhibitor Advanced Gastric Cancer 103
Other antineoplastic agents (belong to one or multiple drug classes)
Arsenic trioxide (Trisenox®) Inhibitor of nuclear factor Kappa-B kinase subunit beta
Thioredoxin reductase 1, cytoplasmic
Transcription factor AP-1
G1/S-specific cyclin-D1
Mitogen-activated protein kinase 3
Mitogen-activated protein kinase 1 inducer
RAC-alpha serine/threonine-protein kinase
Cyclin-dependent kinase inhibitor 1
Histone deacetylase 1
Protein PML
Inducer
Inhibitor
Inducer
Antagonist
Inducer
Inducer
Inducer
Not available
Not available
Not available
Refractory Acute Promyelocytic Leukemia 300
Hydroxyurea Ribonucleoside-diphosphate reductase large subunit Inhibitor Melanoma, resistant chronic myelocytic leukemia, and recurrent, metastatic, or inoperable carcinoma of the ovary and Sickle-cell anemia 301
Paclitaxel (Taxol®) Tubulin beta-1 chain
Apoptosis regulator Bcl-2
Microtubule-associated protein 4
Microtubule-associated protein 2
Microtubule-associated protein tau
Nuclear receptor subfamily 1 group I member 2
Inhibitor
Inhibitor
Not Available
Not Available
Not Available
Inducer
Kaposi's sarcoma and cancer of the lung, ovarian, and breast 302
Docetaxel (Taxotere®) Tubulin beta-1 chain
Apoptosis regulator Bcl-2
Microtubule-associated protein 2
Microtubule-associated protein 4
Microtubule-associated protein tau
Nuclear receptor subfamily 1 group I member 2
Not available
Not available
Not available
Not available
Not available
Binder
Locally advanced or metastatic breast cancer, locally advanced or metastatic non-small cell lung cancer, hormone refractory metastatic prostate cancer, gastric adenocarcinoma and head and neck cancer 303
Ixabepilone (Ixempra®) Tubulin beta-3 chain Inhibitor Breast cancer, head and neck cancer, melanoma, lung cancer, non-Hodgkin's lymphoma, prostate cancer, renal cell carcinoma 279
Methotrexate Dihydrofolate reductase
Humans
Thymidylate synthase
Bifunctional purine biosynthesis protein PURH
Inhibitor
Inhibitor
Inhibitor
Pediatric acute lymphoblastic leukemia, gestational choriocarcinoma, chorioadenoma destruens, breast cancer, epidermoid cancer of the head and neck, lung cancer, and advanced non-Hodgkin's lymphoma 304
Pemetrexed (Alimta®) Thymidylate synthase
Bifunctional purine biosynthesis protein PURH
Dihydrofolate reductase
Trifunctional purine biosynthetic protein adenosine-3
Inhibitor
Inhibitor
Inhibitor
Inhibitor
Malignant pleural mesothelioma, and metastatic non-small cell lung cancer (NSCLC) 305
Streptozocin DNA
Solute carrier family 2, facilitated glucose transporter member 2
O-GlcNAcase BT_4395
Bifunctional protein NCOAT
Cross-linking/alkylation
Ligand
Antagonist
Not available
Malignant neoplasms of pancreas 306
Vinblastine Tubulin alpha-1A chain
Tubulin beta chain
Tubulin delta chain
Tubulin gamma-1 chain
Tubulin epsilon
Transcription factor AP-1
Adduct
Adduct
Adduct
Adduct
Adduct
Other/unknown
Breast cancer, testicular cancer, lymphomas, neuroblastoma, Hodgkin's and non-Hodgkin's lymphomas and Kaposi's sarcoma 307
Vincristine Tubulin beta-1 chain Inhibitor Acute lymphocytic leukemia (ALL), Hodgkin lymphoma, non-Hodgkin's lymphomas, Wilms' tumor, neuroblastoma, rhabdomyosarcoma, relapsed Philadelphia chromosome-negative (Ph-) acute lymphoblastic leukemia (ALL) 308
Vindesine Tubulin beta-1 chain Inhibitor Acute leukaemia, malignant lymphoma, Hodgkin's disease 309
Vinorelbine Tubulin beta chain Antagonist inhibitor Advanced non-small cell lung cancer (NSCLC), relapsed or refractory Hodgkin lymphoma, metastatic squamous cell head and neck cancer, recurrent ovarian cancer, metastatic breast cancer 310

Alkylating agents

One major class of the current drugs belongs to alkylating agents, which can interfere with the formation/linkage of DNA double strands.91 These drugs can cause it by transferring one alkyl group to the guanidine base in DNA (Fig. 2). Cross-linking of nucleic acids with proteins or peptides can likewise affect the DNA geometry, cause erroneous base pairing and DNA strand breakage, and eventually preventing the cell division and causing irreversible senescence.92 Mechanistically, alkylating agents in their electrophilic forms interact with cellular DNA and form covalent adducts, underlying their broader utility. Therefore, alkylating drugs are the frontline chemotherapeutic agents given against most of the cancer types, yet the greatest therapeutic value of their inhibition is known for slow-growing cancers. In Table 1, we described the common chemotherapeutic alkylating agents including the altretamine, chlorambucil, platinum drugs (cisplatin, carboplatin, oxaliplatin), doxorubicin, epirubicin, etoposide, mitoxantrone, cyclophosphamide, thiotepa, and busulfan that are in clinical practice and also provided details of their molecular targets, mode of action, and studied model systems in various malignancies/cancer-types.

Antimetabolites

Antimetabolites are the next class of substances that may compete, replace, or inhibit some specific metabolites inside the cells and thus meddle with the cellular metabolism (Fig. 2).93 These molecules mostly possess a structure similar to a cellular metabolite or enzyme–substrate, usually identified and processed by an enzyme to meet the cellular needs.94 Tetrahydrofolates are formed from unreduced dietary folates by the enzyme dihydrofolate reductase. For example, various medications, such as aminopterin, methotrexate (amethopterin), pyrimethamine, trimethoprim, and triamterene, act directly on the production of these metabolites, hindering the enzyme from producing folate insufficiency.95 Methotrexate, an antifolate drug, is one of the exceptionally successful anti-cancer medications that represses dihydrofolate reductase (DHFR) and obstructs the transformation of dihydrofolic acid (DHFA) into tetrahydrofolic acid (THFA).96 This coenzyme activity is indispensable to the cells as they are essential for the nucleotide synthesis pathways. Methotrexate is an anti-inflammatory molecule and is hence used as a medication for many other inflammatory diseases, like rheumatoid arthritis and extreme psoriasis, apart from various cancer types. A detailed description of common antimetabolites is comprehensively provided in Table 1.

Pyrimidine-derived antimetabolites (pyrimidine antagonists) and Vinca alkaloids

Pyrimidine-derived antimetabolites are a group of chemotherapeutic agents that interrupt DNA synthesis and exert their cytotoxicity to cancer cells (Fig. 2). For instance, cytarabine also known as cytosine arabinoside or Ara C interrupts DNA as well as RNA synthesis by displacing the cytosine with itself (that differs merely by its base sugar arabinose). Cytarabine/cytosine arabinoside is enrolled frequently in clinics to treat leukemia or non-Hodgkin's lymphoma.97 Some other notably pyrimidine-derived antimetabolites include fluorouracil, 5-fluorouracil (5-FU), capecitabine, floxuridine, gemcitabine, decitabine, raltitrexed, and tegafur (Table 1).98, 99, 100, 101, 102, 103 Being as pyrimidine or purine analogs having altered chemical groups, these drugs/compounds induce apoptosis while progressing through the S phase of the cell cycle by their misincorporation to RNA and DNA or by inhibiting the key enzymes required for nucleic acid synthesis. Among these enzymes, DNA polymerases, ribonucleotide reductase, and thymidylate synthetase are the main intracellular targets of the pyrimidine antimetabolites. 5-FU has been a promising pyrimidine analog that largely interrupts DNA and RNA synthesis. 5-FU that dependents on its incorporation to DNA inhibits mitosis and stimulates cell death in the dividing cells.

Vinca alkaloids, primarily extracted from several species under the Vinca genus, are mainly cell division inhibitors, which can interact with microtubular protein tubulin to obstruct its polymerization and meddle with the cytoskeleton, leading to stalling of mitotic division.104 The chromosomes do not separate efficiently during cell division, leading to metaphase capture. Vincristine and vinblastine are primary examples of this drug class with different degrees and ranges of anti-tumor activity (Table 1).105,106 Cancers treated with these drugs include acute leukemia, Hodgkin's and non-Hodgkin's lymphoma, rhabdomyosarcoma, Ewing's sarcoma, neuroblastoma, Wilms' tumor, miscellaneous myeloma, chronic leukemia, thyroid cancer, brain tumors, and several blood-related disorders.107

Other antineoplastic drugs/agents

Besides these three established classes, several other antineoplastic agents are also enrolled in the clinical practice of cancer chemotherapy (Table 1). Given their clinical promises, in the subsequent section, we described the biological activities, targets, and mechanisms of key chemotherapeutic drugs that may belong to one or multiple drug classes and can affect more than one cellular pathway in the cancer cells and induce apoptosis by different mechanisms (Fig. 2). A succinct review of well-known anti-cancer drugs that are in routine clinical practice is provided as follows.

Paclitaxel/Taxol

It is a complex diterpene taxane ring-containing drug acquired from the bark of the Western yew tree (Taxus brevifolia), which induces cytotoxic activity by a novel system.108 It increases the polymerization of tubulin, thereby inducing a mechanism inverse to that of the aforementioned alkaloids.109 It forestalls the microtubule's structural adjustment and depolymerization, bringing an inherent cytotoxic activity to the paclitaxel due to the significance of tubulin-microtubule dynamics.110 The confirmed antagonistic impacts of paclitaxel are reported on the metastatic ovarian and bosom carcinomas after the disappointment from first-line chemotherapy and backslide cases.111 It has also shown efficacy against the advanced stage tumors of the head and neck disease, small cell lung cancer, oesophageal adenocarcinoma, and prostate cancer (Table 1).112,113 Docetaxel is a more potent relative of paclitaxel with a comparable activity. It has been shown to provide benefits against the bosom and ovarian cancers, which get resistant to first-line chemotherapeutic drugs.114 Unfortunately, many side effects have been reported from the long exposure to these drugs, which mainly include neutropenia, arrhythmia, neuropathy, and cardiovascular breakdown (Table 2).115,116

Table 2.

Table listing the adverse effects of various chemotherapy drugs, affected organs, their reported toxicities/side effects and current stage of their clinical progresses.

Drugs/Compound Affected organ Reported toxicity/side effects (frequency in >30% cases) Current status Reference/Sources
Alkylating agents/drugs
Altretamine Blood Nausea and vomiting, Low blood counts, Peripheral neuropathy Approved 278
Bendamustine Blood Low blood counts, Increase in bilirubin levels Approved, Investigational 311,312
Busulfan Blood Low blood counts, Nausea and vomiting (usually mild with standard doses), Loss of fertility, Diarrhea (usually mild with standard doses), Poor appetite, Mouth sores Approved, Investigational 313,314
Carboplatin Cervix, skin, endometrium, esophagus, head and neck, lung, bladder, thymus, mesothelium, testicle Low blood counts, Nausea and vomiting, Taste changes, Hair loss, Weakness, Blood test abnormalities, abnormal magnesium level Approved 315
Carmustine Brain, skin Nausea and vomiting, Facial flushing, Pain and burning at the injection site, Low blood counts Approved, Investigational 280
Chlorambucil Blood Low blood counts Approved 316,317
Cisplatin Testicle, ovary, head-neck, lung, blood, esophagus, trophoblast Nausea and vomiting, Low blood counts, Kidney toxicity, Ototoxicity, Blood test abnormalities (low magnesium, low calcium, low potassium) Approved 318,319
Cyclophosphamide Blood, ovarian, breast, lung, brain Low blood counts, Hair loss, Nausea and vomiting, Poor appetite, Discoloration of the skin or nails Approved, Investigational 320
Dacarbazine Skin, blood Local pain, burning sensation and irritation at the needle site during the infusion, Low blood counts, Nausea and vomiting, Poor appetite, Elevation of blood liver enzymes Approved, Investigational 321,322
Daunorubicin Blood Pain along the site where the medication was given, Urine may appear red, red-brown, orange or pink from the color of the medication for one to two days after you receive a dose, Low blood counts, Nausea or vomiting, Hair loss on the scalp or elsewhere on the body Approved 323,324
Decitabine Blood Low blood counts, Fatigue, Fever, Nausea, Cough, Petechiae, Diarrhea, Constipation, Hyperglycemia Approved, Investigational 325
Doxorubicin Blood, breast, ovarian, bladder, stomach, bronchus Pain along the site where the medication was given, Nausea or vomiting, Low blood counts, Hair loss on the scalp or elsewhere on the body Approved, Investigational 326,327
Epirubicin Breast Low blood counts, Mouth sores, Hair loss on the scalp or elsewhere on the body, Nausea and vomiting, Fatigue, Amenorrhea Approved 321,328
Etoposide Testicle, lung, blood, brain, ovarian Low white blood cell count, Low platelet, Hair loss, Menopause, Loss of fertility, Nausea and vomiting Approved 122
Idarubicin Blood Pain along the site where the medication was given, Low blood counts, Urine may appear red, red-brown, orange or pink from the color of the medication for one to two days after you receive a dose, Nausea or vomiting, Mouth sores (in the first week after therapy), Hair loss on the scalp or elsewhere on the body, Diarrhea/abdominal cramps Approved 329
Ifosfamide Testicle, blood, bladder, bone, pancreas, cervix, breast, stomach, breast, endometrium, lung, ovary Low white blood cell count, Low platelet count, Hair loss, Nausea and vomiting, Poor appetite, Blood in the urine Approved 330
Irinotecan Colon, lung Diarrhea; two types early and late forms, Early diarrhea accompanied by symptoms runny nose, increased salivation, watery eyes, sweating, Fever, flushing, abdominal cramping, Late diarrhea, Nausea and vomiting, Weakness, Low white blood cell count, Low red blood cell count, Hair loss, Poor appetite, Weight loss Approved, Investigational 331
Lomustine Brain, blood Low blood counts (bone marrow suppression), Nausea and vomiting Approved, Investigational 332
Mechlorethamine or Chlormethine Blood, lung, brain Low blood counts, Nausea and vomiting, Hair loss, Mouth sores, Redness, dryness, irritation with topical use, Loss of fertility Approved, Investigational 15
Melphalan Blood, ovary, breast, skin Low blood counts, Nausea and vomiting Approved 333
Mitomycin C Stomach, intestine, breast, lung, bladder, pancreas, crevice Low blood counts, Mouth sores, Poor appetite, Fatigue Approved 137
Mitoxantrone Prostate, blood, breast Low blood counts, Nausea and vomiting, Fever, increases in blood tests measuring liver function Approved, Investigational 334
Oxaliplatin Intestine, pancreas, stomach Peripheral neuropathy, Nausea and vomiting, Diarrhea, Mouth sores, Low blood counts, Fatigue, Loss of appetite Approved, Investigational 335,336
Temozolomide Brain, skin Nausea and vomiting, Constipation, Headache, Fatigue Approved, Investigational 337
Topotecan Ovary, lung, cervice Low blood counts, Nausea and vomiting, Hair loss, Diarrhea Approved, Investigational 338, 339, 340
Thiotepa Breast, ovary, thyroid, bladder Low blood counts, Hair loss Approved, Investigational 341
Trabectedin Ovary Anemia, Increased liver enzymes, Low white blood cell count, Nausea, Low platelets, Vomiting, Fatigue, Low potassium, Low phosphorous, Decreased appetite, Constipation, Increased CPK Approved, Investigational 342,290
Purine antimetabolites (purine antagonists)
Azathioprine Blood Black, tarry stools, Bleeding gums, Blood in the urine or stools, Chest pain, Chills, Cough, Fever, Hoarseness, Lower back or side pain, Painful or difficult urination, Pinpoint red spots on the skin, Sore throat, Sores, ulcers, or white spots on the lips or in the mouth, Swollen glands, unusual bleeding or bruising, unusual tiredness or weakness Approved 291
Cladribine Blood Fever, Fatigue, Low white blood cell count, Low red blood cell count, Neutropenic fever, Myelosuppression Approved, Investigational 343
Clofarabine Blood Leukopenia, Anemia, Infection, Limb pain, Lymphopenia, Low platelet count, Elevated liver enzymes, Vomiting, Nausea, Itching, Neutropenia, Diarrhea, Neutropenic fever, Creatinine increased, Bilirubin increased, Chills, Headache, Fever, Rash, High heart rate, Abdominal pain, Fatigue, Decreased appetite Approved, Investigational 293,344
Fludarabine Blood Low blood counts, Fever, Infection, Weakness, Cough, Nausea and vomiting, Poor appetite Approved 294
Gemcitabine Breast, ovary, lung, pancreas, bladder Flu-like symptoms (muscle pain, fever, headache, chills, fatigue), Fever (within 6–12 h of first dose), Fatigue, Nausea (mild), Vomiting, Poor appetite, Skin rash, Low blood counts, Temporary increases in liver enzymes, Blood or protein in the urine Approved, Investigational 295
Mercaptopurine Blood, intestine Low blood counts, Liver toxicity, Increased bilirubin, Increased liver enzymes, jaundice, abdominal swelling (ascites) Approved 345
Nelarabine Blood Low blood counts, also known as bone marrow suppression (anemia, Fatigue, neutropenia, thrombocytopenia), Nausea Approved, Investigational 297
Pentostatin Blood Cough or hoarseness, Fever or chills,
Lower back or side pain, Painful or difficult urination, Skin rash or itching (sudden), Unusual tiredness or weakness
Approved, Investigational 346
Thioguanine Blood Low blood counts Approved 347
Pyrimidine antimetabolites (pyrimidine antagonists)
Capecitabine Breast, stomach, intestine Low white blood cell count, Low red blood cell count, Hand-foot syndrome, Diarrhea, Elevated liver enzymes, Fatigue, Nausea and vomiting, Rash and itching, Abdominal pain Approved, Investigational 99
Cytarabine Blood Headache, Low blood counts, nausea and vomiting, Mouth sores, increases in blood tests measuring liver function Approved, Investigational 97
Gemcitabine Breast, ovarian, lung, pancreas, bladder Flu-like, Fever, Fatigue, Nausea, Vomiting, Poor appetite, Skin rash, Low blood counts, Temporary increases in liver enzymes, Blood or protein in the urine Approved 348,349
Fluorouracil Breast, colon, pancreas, skin, stomach, rectal Diarrhea, Nausea and possible occasional vomiting, Mouth sores, Poor appetite, Watery eyes, sensitivity to light, Taste changes, metallic taste in mouth during infusion, Low blood counts, white and red blood cells and platelets may temporarily decrease, Skin reactions, Hair thinning, Hand -foot syndrome Approved 98
Raltitrexed Colon, mesothelium Increased risk of infection, Breathlessness and looking pale, Tiredness and weakness during and after treatment, Diarrhoea or constipation, Feeling or being sick, Skin problems, Mouth sores and ulcers, Tummy (abdominal) pain, Liver changes, Loss of appetite and weight loss, Dehydration Approved, Investigational 102
Tegafur Stomach Myelosuppression, Central neurotoxicity, Gastrointestinal toxicity Approved, Investigational 350
Other antineoplastic agents (belong to one or multiple drug classes)
Arsenic trioxide (Trisenox®) Blood Nausea and vomiting, Cough, Fatigue, Fever, Headache, Rapid heartbeat, Sore throat, Abdominal pain, Diarrhea, Blood test abnormalities (low potassium and magnesium), Shortness of breath, Blurred vision and eye irritation, elevated blood sugar level, swelling of the face, hands, feet or legs, Difficulty sleeping, Rash, Heart rhythm changes, Joint pain, Itching, Numbness or tingling of hands or feet Approved 351,352
Hydroxyurea Skin, blood, ovary, breast Low blood counts Approved 301
Paclitaxel (Taxol®) Bone, lung, ovary, breast Low blood counts, Hair loss, Arthralgias and myalgias, pain in the joints and muscles, Peripheral neuropathy, Nausea and vomiting, Diarrhea, Mouth sores, Hypersensitivity reaction Approved, Vet Approved 353
Docetaxel (Taxotere®) Breast, lung, prostate, stomach Low white blood cell count, Low red blood cell count, Fluid retention with weight gain, swelling of the ankles or abdominal area, Peripheral neuropathy, Nausea, Diarrhea, Mouth sores, Hair loss, Fatigue and weakness, Infection, Nail changes Approved, Investigational 296
Ixabepilone (Ixempra®) Breast, skin, lung, blood, prostate, kidney Peripheral neuropathy, Weakness, Muscle and joint pains, Hair loss, Nausea and Vomiting, Low white blood cell count Approved, Investigational 354
Methotrexate Blood, breast chromium Dizziness, Drowsiness, Headache, Hair Loss, Swollen, Tender Gums Decreased Appetite, Reddened Eyes Approved 355
Pemetrexed (Alimta®) Pleura, lung Fatigue, Nausea Approved, Investigational 305
Streptozocin or Streptozotocin Pancreas Nausea and vomiting, Nephrotoxicity Approved, Investigational 356
Vinblastine Breast, blood, testicle, bone Low blood counts, Injection site reactions, Fatigue and weakness Approved 307
Vincristine Blood Hair loss, neurotoxicity, alopecia, myelosuppression Approved, Investigational 357
Vindesine or Eldisine Blood Myelosuppression, Neurotoxicity Approved, Investigational 358
Vinorelbine Lung, blood, ovary, breast Low blood counts, Nausea or vomiting, Muscle weakness, Constipation Approved, Investigational 359,360

Etoposide

Etoposide is a semi-synthetic derivative of a plant glycoside called podophyllotoxin.117,118 It causes DNA damage by altering DNA topoisomerase-II function, and posing G2 cell cycle arrest.119 Etoposide creates a ternary complex with DNA and the topoisomerase–II complex, preventing the latter's binding to the DNA strands, while in doing so, it causes damage to DNA strands.120 Cancer cells depend on this enzyme more than healthy cells, as they divide more quickly; therefore, it causes defects in DNA synthesis that promotes apoptosis of the cancer cells.121 Etoposide is effective against several aggressive malignancies including Kaposi's sarcoma, Ewing's sarcoma, testicular disease, lung disease, lymphoma, non-lymphocytic leukemia, and glioblastoma multiforme (Table 1).122

Camptothecin and its derivatives

Camptothecin is a long-known topoisomerase inhibitory compound of natural origin. It was initially extracted from the bark of the tree Camptotheca acuminata.123,124 Camptothecin is a cytotoxic alkaloid comprised of a pentacyclic ring structure containing a pyrrole (3,4-β) quinoline moiety, an S-configured alpha-hydroxy lactone form, and a carboxylate form.125 In later years, many more effective derivatives of this topoisomerase inhibitor have been synthesized and are effectively being applied in cancer chemotherapy.126 The most recognized drug of this class, topotecan, is used against the advanced cancers of the ovary and lung. It targets topoisomerase-I, an enzyme responsible for easing the torsional strain in DNA by inducing single-strand breaks during replication.120 The inhibition of topoisomerase-I by the active compounds may lead to sudden inhibition of DNA replication and transcription, causing growth arrest (Table 1).127 In the physiological conditions, topotecan remains in equilibrium with its carboxylate structure.128 Its active lactone can intercalate between DNA bases in the topoisomerase-I cleavage complex.129 The coupling site of topotecan in the cleavage complex interferes with the topoisomerase-I while reiterating the scratched DNA strand in the wake of releasing the strain. Such intercalation traps the topoisomerase-I in the cleavage complex bound to the DNA.130 Another well-known drug of this class is irinotecan that has shown efficacy against late-stage colorectal carcinoma, aggressive growths in cancers of the lung, cervix, ovary, colon, etc.131,132

Actinomycin D (dactinomycin)

A transcription inhibitor chromopeptide molecule actinomycin D (also called dactinomycin) binds to DNA while initiating transcription at initiation complex and inhibits RNA elongation steps.133 It can intercalate, preferably between guanidine and cytosine, or bind the DNA strand's minor grooves to form a highly stable drug-DNA complex, preventing DNA unwinding and thus stalling the progress of the RNA chain.134 Dactinomycin was initially obtained from Streptomyces sp., identified as an antibiotic agent, but later its anti-tumor potential was recognized, and thus it was inducted in the chemotherapy as an anticancer antibiotic to treat Ewing's sarcoma, Wilms' tumor, rhabdomyosarcoma, trophoblastic neoplasm, testicular and ovarian cancers, etc (Table 1).135

Mitomycin C

Mitomycins, a class of natural quinones, are also derived from Streptomyces caespitosus and are known for their transformation and competence, enhancing potential in bacterial cells. It is also a known antineoplastic agent as it slows down fibroblast cell growth in glaucoma.136,137 These bioreductive alkylating agents (especially mitomycin C derivatives) are also utilized as anti-cancer therapeutics because their structural modification impedes the cell cycle during G1 and S phase transitions. Its reduction to form mitosene can lead to the N-alkylation of DNA bases.138 It can lead to inhibition of replication by cross-linking at adenine N6 and guanine O6 and N2.139 It is effectively used in the palliative treatment of non-small cell lung carcinoma and other solid tumors of breast, colorectal, oesophageal, pancreatic, and cervical origins. Porfiromycin is an N-methyl derivative of mitomycin C that is found effective against cancer of head and neck regions (Table 1).140

Daunorubicin (rubidomycin)

Daunorubicin causes a break in DNA strands by actuating topoisomerase-II and producing quinone-type free radicals. Daunorubicin, otherwise called daunomycin, is a chemotherapy medicine used to treat cancer growth in acute myeloid leukemia (AML), acute lymphocytic leukemia (ALL), chronic myelogenous leukemia (CML), and Kaposi's sarcoma, etc.141,142 It is applied by infusion into a vein, and following its intercalation with DNA, it hinders macromolecular biosynthesis. This predominantly obstructs the movement of the enzyme topoisomerase-II, which loosens up the DNA supercoiling.143 Daunorubicin impedes the topoisomerase–II complex after it unties the DNA chain for replication and thereby it represses the DNA double helix from resealing and therefore interferes with the replication to proceed (Table 1).144,145

Bleomycin

Another Streptomyces compound with anti-tumor activity bleomycin is mostly used against Hodgkin's lymphoma, ovarian and testicular cancers. It chelates copper or iron, produces superoxide radicals, and intercalates between DNA strands – causes chain scission.146 Emerging evidence proposes that bleomycin also hinders the consolidation of thymidine into DNA strands, and the cleavage of DNA relies upon oxygen and metal particles in vitro.147 The specific mechanism of DNA strand scission is uncertain, yet it has been proposed that bleomycin chelates metal particles (principally iron), delivering a pseudoenzyme that responds with oxygen to create superoxide and hydroxide free radicals that divide DNA; however, little myelosuppression is the unique highlight.148 The drug has also been effective against squamous cell carcinoma of the skin, and cancers of the mouth, head, neck, genitourinary tract, and throat (Table 1).

Lateral damage: Putting multiple organs at risk

Aggressive tumors are primarily marked with uncontrolled and extensive cell division that may compromise the normal physiology of other non-transformed cells in the vicinity of affected organs/tissues. In many ways, cancer cells achieve super-specialized metabolic and survival potential with the capacity to tolerate extreme stresses like low oxygen and nutrient supplies. Cancers might start from mutation of one type or the other, but as the disease progresses,149,150 multiple mutations and genetic abnormalities accumulate that lead to increased tumor heterogeneity.87 The heterogeneous nature of tumor mass plays a vital role in developing resistance against most of the drugs that we have described here. This leads to a challenge of its kind, leaving doctors with the only option of changing the medication after certain intervals as one type of drug stops acting against tumors. In many cases, the combination therapy that includes more than one drug at a time could be an effective strategy against some form/type of cancers, such as cancers affecting bone marrow (leukemia) and lymph nodes (lymphoma). Clinical trials help a lot in understanding the effects of new combinations and thus have rapidly improved the cancer chemotherapy paradigm.

The sustainability of chemotherapy relies upon its continuous effect on cell division and tumor growth. As discussed above, the majority of the treatments work by compromising the stability or integrity of nucleic acids that tend to stall the cell cycle or activate programmed cell death pathways. Other drugs may act by generating extreme proteotoxic stress inside the cells by interfering with the protein folding or degradation machinery, including chaperone, proteasome, and autophagy.151, 152, 153, 154 The idea is to generate cellular stresses so that the cancer cells that are already under compromised physiology may die while other cells can survive. However, it is evident from the above descriptions that most of these drugs act by interfering with one or multiple cellular pathways of extreme importance.155 Therefore, chemotherapeutic drugs are mostly referred to as cytotoxic agents, causing a plethora of side effects. In most cases, long-term treatments may lead to enormous toxicity causing severe damage or failure of multiple organs. This may lead to enormous physiological and psychological stress to the patients under chemotherapeutic medication. Table 2 summarizes the adverse effects of various chemotherapeutic agents and the current stage of their clinical progress. The side effects of these drugs may depend on many factors and variables, such as type, stage, and location of the tumor, along with age, health, and other clinical aspects of the patients. The most common and readily visible side effects originate from the gut that may lead to nausea, vomiting, constipation, and diarrhea.156,157 Most of the anticancer drugs can directly interrupt the cell division pathways; therefore, the first line of affected organs may also include those which are formed with continuously dividing cells, such as skin, hair, and intestinal linings.158,148 Skin rashes, alopecia, and mucositis are the most visible side effects that one can observe in patients undergoing chemotherapy.157,159 Radiation also contributes significantly to these debilitating changes. A general problem observed in many patients is fatigue that can occur due to multiple factors, including depression, anemia, high doses of radiation therapy, etc.160

Chemotherapeutic drugs may also directly injure the bone marrow and cause a steep decline in blood cell count, exposing patients to several other types of possible infections.161 Thrombocytopenia, a condition of excessive bleeding because of lower platelet numbers, is observed more frequently in patients taking alkylating and/or antimetabolites agents.162 Alkylating agents may further trigger conditions, such as hemorrhagic cystitis leading to microscopic haematuria, nocturia, dysuria, etc. along with mild or intense suprapubic pain.163 Whereas, antimetabolites can generate enormous toxicity in vital organs, like the liver and kidneys.164,165 The most devastating of all is their direct impact on cardiovascular health. Different classes of chemotherapeutic drugs have varying impacts on cardiac cells, leading to multiple types of systemic conditions adversely affecting patients' survival.166 Loss of libido and sexual dysfunctions are other conditions caused due to long-term exposure to these drugs, which affects not only physical well-being but also the patient's psychological health (Table 2).167,168

These drugs and radiation therapies may have several direct or indirect impacts on cognitive functioning leading to a condition, which is now commonly referred to as chemo brain.169,170 This is a condition of the inability of someone undergoing chemo-drug therapies to do their routine cognitive functions, which may be related to their memory, cognition, and behavior.171 The drugs may adversely impact communication, reasoning, learning, problem-solving, memory storage, concentration, following commands, etc. Damage of optic and the ocular motor nerves is also observed in several patients taking antineoplastic drugs that may lead to the weakening of vision as well.172,173 The majority of chemotherapeutic agents affect these tissues in a dose-dependent manner; though, there are differences in their sensitivity to individuals due to enormous factors such as metabolism efficiency, genetic makeup, other medication, and environmental factors (Table 2).174 Despite enormous toxicities and side effects prevalent in the public domain, the chemotherapy medicines remained the most immediate and viable therapeutic option in most cases, where we can see the cancer cells but cannot remove them through surgery or source of radiation. To address various above-described detrimental physical and psychological changes, modern-day clinical practices involve a comprehensive team of doctors and specialists for the patients who are undergoing long-term treatments and are more prone to develop severe physiological complications.

Recent advancements and next-generation therapeutic approaches

Since the early 19th century, rigorous investigations and clinical studies were carried out around the world. Several research groups have made landmark discoveries during and after the Second World War. Following the development of a fundamental description of the disease and associated pathways, the focus shifted majorly towards advancing the diagnosis and treatment methods of the disease. Despite enormous progress being made in the second half of the last century, not much could have been achieved in successfully curing most of the cancers. One primary reason behind the failures is our inability to diagnose the oncogenic changes inside the body at the early stages of cancer development. In recent years, the concept of liquid biopsies has emerged that primarily relies on the screening of mutated DNAs, proteins, RNAs, and other genetic markers in the patient's blood samples.175, 176, 177 This practice can help diagnose the disease at a much earlier stage than we do now, using the traditional tissue biopsies and imaging methods.178 New technological tools, like advanced software and robotic platforms, have also speeded cancer diagnosis and treatment. In this subsequent section taking chemotherapy into account, we compressively reviewed the recent progress made in the targeted drug delivery, personalized medication, and immunotherapy (Fig. 3).

Figure 3.

Fig. 3

Schematic diagram showing targeted cancer therapeutic approaches. (A) Model showing targeted drug delivery approach of anti-cancer drugs/chemical agents conjugated with receptor substrate specific to the cancer cell receptor. (B) Model showing the building of a personalized medicine approach based on omics data and drug response study targeting specific mutation/signature in individual patients for precision cancer therapeutics. (C) Model showing immunotherapeutic regime involving reprogramming of T-cells to target cancer cells by a chimeric antigen receptor for their application in cancer immunotherapy.

Targeted drug delivery

The major drawback of most chemotherapeutic treatment plans is their non-specificity or inability to ascertain and target cancerous cells directly. Therefore, this lack of specificity poses a major therapeutic limit reflected in the failure of delivering drugs locally to the cancer tissue mass. In the past few decades, many new approaches have been constituted and tested, and have given hopes for future drug delivery (Fig. 3A). Nanoparticle-based delivery methods, targeted antibodies, aptamer functionalization, and specific drugs like Herceptin (in breast cancer) offered the promise that can harness great success in the upcoming years.179, 180, 181 Cancer cell-directed antibodies are at the forefront of targeted drug delivery methods and have accelerated the hunt for more effective or potentially improvised approaches.182,183 Different types of nanocarriers and formulations of nano-drugs have also improved the delivery of drug compounds to the cancer cells.184, 185, 186 The most significant advantage of targeted drug deliveries is that the enormous drug-mediated toxicities on other adjacent tissues and organs can be minimized. We can also avoid most of the collateral damage leading to stress and organ failures. However, the effectiveness of many of these delivery methods in patients is still under clinical evaluation. Further details regarding these delivery systems can be accessed in the previously published reports.187,188

Personalized medication

Given the heterogeneity of tumor cells, the cancer of one patient is different from that of another in terms of accumulated genetic abnormalities, its pace and progression, the response to the treatment, and the chances of developing resistance to the administered drugs.45 The current technologies have made it possible to get insights into this disease heterogeneity or explore molecular variations at the individual level, thereby tailoring the treatment regime as per the personalized needs of every cancer patient.149,189 This helps in choosing the most appropriate drugs and their dosages for a better outcome in cancer management.190 For instance, dabrafenib and vemurafenib are directed against BRAF gene mutation, while trastuzumab works well against HER-2 mutated cells.47,191,192 The idea here is to dive deep into the omics (genomic, transcriptomic, proteomic, metabolomic, etc.) data of the patient to identify the major molecular players fostering the transformed cells, and design the treatments against these clinically actionable targets (Fig. 3B).155,193,194 Different types of mutations have been observed to be frequent in specific subtypes of cancer. Therefore, measuring and manipulating these driver mutations based on an individual patient is expected to yield better results. Following are some examples of drugs that are potentially effective in patients harbouring some specific type of genomic alteration. A mutation in anaplastic lymphoma kinase (ALK) was reported to drive tumor formation in 5% of the non-small-cell lung cancer population and led to the discovery of ALK blockers such as crizotinib and ceritinib.195 An oral BRAF inhibitor, namely vemurafenib has been shown to possess a greater affinity for the mutant BRAFV600E than the wildtype BRAF, and therefore was recommended to the patients carrying the above mutation in the tumors.196 Several potential targets for novel drugs have been identified using high-throughput screening technologies, and various compounds against them have been recently approved or are under investigation. Similarly, alterations in phosphoinositide 3-kinases (PI3K)/protein kinase B (AKT)/mTOR pathway have also been frequently found in many solid tumors. Among these, activation of phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) and loss of phosphatase and tensin homolog (PTEN) mutations are the most common. Basket trials have been conducted to evaluate the effectiveness of adding alpelisib, a specific p110α PIK3CA inhibitor to the hormonal treatment for PIK3CA mutant luminal breast cancer patients. The results emphasize the importance of checking for this molecular alteration in patients and following the tested treatment regime for better anti-tumor response.193 Next, aberrant changes in the fibroblast growth factors (FGF) signaling pathway have also been associated with the initiation and progression of oncogenic events. Different types of genetic alterations in FGF receptor amplification, mutations, and gene-fusion have been shown to respond variably to certain drugs, and hence demand molecular profiling for the selection of effective drugs.46,197 Another promising example is of using poly ADP ribose polymerase (PARP) inhibitor olaparib for the treatment of breast cancer susceptibility gene (BRCA)-mutant cancers for better targeting of cancer cells, without much effect on the normal cells.198,199 On the cytoprotective side, recently a natural alkylated withanolide viz. 2,3-Dihydro-3beta-methoxy withaferin-A was shown to protect normal cells against a variety of stresses enrolled in cancer therapeutics.200 The idea of precision or personalized medicine is new but has been beneficial for lots of patients; however, the cost of these new generation approaches for genome profiling is still high, making it unaffordable for a large population of the world.201

Antimicrobial peptides

Advances in cancer therapy have prompted scientists to look for natural tumoricidal chemicals, such as antimicrobial peptides (AMPs), that could be used for cancer treatment.202, 203, 204 In addition to antibacterial action, several AMPs have been shown to have intrinsic anticancer potential.205,206 These molecules constitute an intriguing new resource for anticancer drugs, and they provide several distinct therapeutic advantages over other anticancer treatments since they do not bind directly to any particular receptor in cancer cells.207 The polycationic and amphipathic character of AMPs has been postulated as a possible explanation for the preferential interaction of AMPs with cancer cells, as compared to normal cells.208,209 Membrane disruption has been identified as a critical stage in the microbial killing mechanism of several AMPs. These anticancer effects of AMPs are unaffected by molecular heterogeneity inside a tumor or across tumors, nor are they counteracted by cancer cells' flexibility and alternate survival pathways, as is commonly found in cancer resistance (Fig. 4).204,209,210

Figure 4.

Fig. 4

Advantages of antimicrobial peptides as anti-tumor agents.

Cathelicidins are members of the antimicrobial peptide family found in granules of vertebrate neutrophils.211 The only human cathelicidin is the cationic peptide LL-37. Cathelicidins are composed of a highly conserved N-terminal signal peptide including a cathelin domain and a C-terminal cationic antimicrobial peptide. Apart from their antimicrobial activity, cathelicidin-derived antimicrobial peptides exhibit a range of biological roles, including antiviral, antifungal, anticancer, and immunomodulatory properties.212,213 LL-37 exhibits anticancer activity against oral cancer cells and has been identified as a molecular target for colon cancer.214 Immunohistochemical staining of colon cancer tissue showed that LL-37 is abundantly expressed in normal colon mucosa but downregulated in colon cancer tissues due to DNA methylation in the LL-37 promoter.215

In human oral squamous cell carcinoma SAS-H1 cells, the active domain of LL-37 induces mitochondrial depolarization, which results in caspase-independent apoptosis. Furthermore, mice lacking cathelicidin are more susceptible to azoxymethane-induced colon carcinogenesis.216

Many important advancements have been achieved using antimicrobial peptides against colorectal cancer.217 Using LL-37 on the surface of magnetic nanoparticles led to a significant reduction in cell viability of colon cancer cell lines, inducing apoptosis at a much higher rate than in the case of LL-37 peptide alone.218

Fan et al created a biodegradable and injectable thermosensitive hydrogel containing docetaxel and LL-37 polymeric nanoparticles. Intraperitoneal injection of this hydrogel substantially inhibited the development of HCT116 carcinomatosis in a mouse model of colorectal carcinoma.219

In comparison to existing chemotherapeutic agents, the flexible nature of peptide structures, along with non-receptor activity, might confer strategic benefits on AMPs for the treatment of colon cancer in the future.220 New treatment approaches for patients with advanced-stage, unresectable melanomas continue to be of significant clinical interest, as these patients cannot be treated surgically and have only a modest or temporary response to chemotherapy and radiation. In phase 1 clinical trial NCT02225366, intra-tumoral injections of LL-37 are being explored for melanoma patients with cutaneous metastases to determine the most effective dose of LL-37 that could be administered. Patients will receive weekly intra-tumoral injections of LL-37 (starting dose, 250 μg/tumor) every 7 days for up to 8 weeks. Researchers are also interested in understanding whether LL37 can activate the immune system, which could help in the control of the disease.221

Although AMPs have the potential to develop into a more effective and safer alternative to conventional cancer therapy, future research and development of anticancer peptide therapeutics will most likely focus on the poor pharmacokinetics, the control of selectivity, toxicity, and routes of administration through the establishment of appropriate in vivo models.222

Immunotherapy

Supplementing our immune system by devising immunotherapy strategies is another highly-specific and largely promising approach to address the problem of late-stage cancer therapeutics.223,224 Several new approaches have been developed in the last decade to modify and train a patient's immune system to search, identify and target the transformed cells inside the body by recognizing multiple cell surface markers.225 For instance, supplementing the treatment regime with immunotherapy has significantly improved the success rate of the chemotherapeutic drugs in many cancer types, especially the triple-negative breast cancers.226, 227, 228 The immunotherapy drugs are given alone or in combination with chemotherapy drugs to achieve better clinical outcomes.229 Prophylactic and therapeutic vaccines have been developed to help patients' immune systems towards learning to tackle the transformed cells.230 Atezolizumab is an example of an anti-cancer immune drug that helps to dissolve hard-to-treat breast cancers.231, 232, 233 Tisagenlecleucel, an FDA-approved drug, treats B-cell lymphoblastic leukemia by effectively engineering the chimeric T cell receptor (CAR-T) of the patient (Fig. 3C).234 Some treatment plans also include interferons and interleukins to jump-start the fight against cancer cells by boosting the patient's immune system. Moreover, the field of immunotherapy is gradually evolving and is beyond the scope of our present article.

Combination therapy

Combination therapy may offer the advantages of the fast tumor regression of targeted therapies with the long-lasting responses induced by immunotherapy.235 Immunotherapy may be able to consolidate the dramatic tumor responses achieved with targeted therapy into durable, long-lasting remissions, in which sustained host responses targeting multiple antigens may reduce the risk of developing potentially lethal drug-resistant clones.236 Combining cancer vaccines with immune checkpoint inhibitors (ICIs) such as those targeting cytotoxic T lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1/programmed cell death ligand 1 (PD-1/PD-L1) is one approach to avoid T cell anergy.237,238 Moreover, T-cell and NK-cell invasion may be facilitated by targeted drugs that modify the tumor endothelium.239 Gemcitabine, for example, is a deoxycytidine analog that induces tumor cell death, which results in antigen cross-presentation and cross-priming.240,241 Carboplatin, pemetrexed, and pembrolizumab are now commonly used in the treatment of non-small cell lung cancer as first-line agents.242 For triple-negative breast cancer, nab-paclitaxel and atezolizumab (anti-PD-L1) were recently authorized based on an improvement in progression-free survival (PFS) over chemotherapy.243

Adenosine receptor antagonists (ARs) are involved in regulating tumor progression.244 The four ARs discovered so far, A1, A2A, A2B and A3, belong to the class A family of G protein-coupled receptors (GPCRs).245 The A2BR seems more promising as a target in chemotherapeutic interventions,246 whereas the A2AR is the main target in the immunotherapeutic approach.247,248 The existence of dual compounds, i.e., molecules acting on the A2AR and on the A2BR, has prompted their use in both immunotherapeutic and chemotherapeutic interventions.249,250

Epigenetic modulators may have a dual function in the tumor microenvironment, boosting both antigen expression and T cell effector activity.251,252 ICI-induced reactivation of exhausted T cells is linked with considerable chromatin remodeling, indicating that epigenetic modification may relieve certain patients with exhausted T cell function.253,254

While many strategies focus on modifying the native tumor microenvironment, an alternative strategy involves the direct infusion of engineered immune cells or cellular receptors into the patient via adoptive cellular therapy with the goal of increasing the number of effector cells that recognize tumor-expressed antigens.255 Upon reinfusion, lymphocytes grown ex vivo may respond more potently to tumor antigen since they have not been continuously exposed to tolerogenic microenvironmental signals.256 Tumor infiltrating lymphocytes (TIL) treatment,257 chimeric antigen receptor (CAR) engineered T-cells,258,259 and TCR-engineered cell therapy260,261 are the three main forms of adoptive T cell therapy currently being investigated.257,262,263

Injecting oncolytic viruses directly into the tumor microenvironment is an alternate technique for improving tumor antigen recognition and strengthening T cell responses.264 Talimogene laherparepvec (T-VEC) is a modified herpes simplex virus that is injected intra-lesionally into the melanoma tumor in individuals who have failed to get their tumor removed. It induces immediate lysis of tumor cells and the production of granulocyte-macrophage colony-stimulating factor (GM-CSF), which serves as a cytokine to stimulate the recruitment, maturation, and activity of antigen presenting cells (APCs).265

Several studies combining immunotherapy with molecularly targeted treatment, either concurrently or sequentially, have shown hepatotoxicity as a major concern.266 Combinations of dabrafenib, trametinib, and anti-PD-1 treatment have been associated with a greater risk of grade 3/4 adverse events in melanoma than would be expected with targeted therapy alone.256

Emerging drugs are beginning to take a more comprehensive perspective of the tumor microenvironment (TME) and are revealing complementary techniques to simultaneously restore and enhance immune activity and promote therapeutic synergy.267,268 Future research of combined therapy should continue to concentrate on exploring the intricate interplay between targeted drugs and immunotherapy, as well as optimizing parameters such as administration time, dose, and sequencing that may enhance the therapeutic index.240

Conclusions and future perspectives

Chemotherapy is a largely investigated and clinically assessed approach in cancer therapeutics. In this report, we summarized the present status of chemotherapy and several drugs/agents presently enrolled in the clinical practice to provide an overview of how the field has evolved over the years and has traced the possibilities and druggability of diverse cancers in the clinic. New trends are evolving in the drug discovery paradigm that includes designing new derivatives of old drugs with high efficacy and lower toxicities.269,270 Several natural compounds isolated from microbial, plants, or natural sources were suggested to hold enormous potential to target stress response pathways by mounting a hyperactive response instigating cancer cell death.271, 272, 273, 274 Natural compounds/agents as chemotherapeutic drugs demonstrated several advantages over synthetic ones, owing to their less cytotoxicity, cost, and straightforward production/extraction process.273,275,276 Therefore, recognition and attenuation of treatment toxicity in cancer chemotherapy will be a key factor in determining the success of its regimes in the future.

Revised clinical strategies further underlined that combination therapy could emerge as a promising therapeutic approach to treat diverse cancers in the future. Combination strategies comprise an obvious overlap of the chemotherapeutic regimes with the targeted drug delivery, personalized medicine, and immunotherapy. It may not only improve the efficacy of enrolled treatment but can also elicit the responsiveness of certain tumors towards their effective therapeutic targeting in the clinic. Among these, personalized combination therapies that target individual tumor types based on their molecular signatures may offer a great promise. With acquiring more personalized insights in the chemotherapeutic treatments, a greater emphasis will be on individual regimes that could meet a high clinical success.277 Therefore, chemotherapy beyond its conventional clinical efficacy offers to improve the clinical outcome in combination therapies. Translation of these combined approaches in the clinics is yet to see a complete success and thus warrants more clinical investigations.

Conclusively, clinical practice of cancer chemotherapy has achieved considerable success, however, it still holds scope to improve, particularly in terms of efficacy and safety of enrolled chemotherapeutic regimes. Therefore, it further requires concerted efforts and careful investigations assessing the efficacy of combined therapeutic regimes. In summary, more investigative focus on drug design in individual or combined regimes and rigor on their clinical trials involving careful patient selection and stratification are warranted to achieve the greater therapeutic success of chemotherapeutic regimens in the clinic.

Conflict of interests

The authors declare no conflict of interests.

Funding

This research was funded by "Agencia Canaria de Investigación, Innovación y Sociedad de la Información (ACIISI) del Gobierno de Canarias” (No. ProID2020010134), and óCaja Canarias (Project No. 2019SP43).

Acknowledgements

All the authors are highly grateful and acknowledge to the authority of the respective departments and institutions for their support in carrying out this research. The authors also express their sincere gratitude to the unknown referee for critically reviewing the manuscript and suggesting useful changes.

Footnotes

Peer review under responsibility of Chongqing Medical University.

Contributor Information

Amarnath Mishra, Email: amishra5@amity.edu.

José M. Pérez de la Lastra, Email: jm.perezdelalastra@csic.es.

References

  • 1.Janssen L.M.E., Ramsay E.E., Logsdon C.D., Overwijk W.W. The immune system in cancer metastasis: friend or foe? J Immunother Cancer. 2017;5(1):79. doi: 10.1186/s40425-017-0283-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Laplagne C., Domagala M., le Naour A., et al. Latest advances in targeting the tumor microenvironment for tumor suppression. Int J Mol Sci. 2019;20(19):4719. doi: 10.3390/ijms20194719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Emon B., Bauer J., Jain Y., Jung B., Saif T. Biophysics of tumor microenvironment and cancer metastasis - a mini review. Comput Struct Biotechnol J. 2018;16:279–287. doi: 10.1016/j.csbj.2018.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Costa A., Scholer-Dahirel A., Mechta-Grigoriou F. The role of reactive oxygen species and metabolism on cancer cells and their microenvironment. Semin Cancer Biol. 2014;25:23–32. doi: 10.1016/j.semcancer.2013.12.007. [DOI] [PubMed] [Google Scholar]
  • 5.Gilbert S., Oliphant C., Hassan S., et al. ATP in the tumour microenvironment drives expression of nfP2X7, a key mediator of cancer cell survival. Oncogene. 2019;38(2):194–208. doi: 10.1038/s41388-018-0426-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Pfirschke C., Siwicki M., Liao H.W., Pittet M.J. Tumor microenvironment: no effector T cells without dendritic cells. Cancer Cell. 2017;31(5):614–615. doi: 10.1016/j.ccell.2017.04.007. [DOI] [PubMed] [Google Scholar]
  • 7.Bedoui S., Herold M.J., Strasser A. Emerging connectivity of programmed cell death pathways and its physiological implications. Nat Rev Mol Cell Biol. 2020;21(11):678–695. doi: 10.1038/s41580-020-0270-8. [DOI] [PubMed] [Google Scholar]
  • 8.Rodrigues D., Souza T., Jennen D.G.J., Lemmens L., Kleinjans J.C.S., de Kok T.M. Drug-induced gene expression profile changes in relation to intestinal toxicity: state-of-the-art and new approaches. Cancer Treat Rev. 2019;77:57–66. doi: 10.1016/j.ctrv.2019.06.004. [DOI] [PubMed] [Google Scholar]
  • 9.Delou J.M.A., Souza A.S.O., Souza L.C.M., Borges H.L. Highlights in resistance mechanism pathways for combination therapy. Cells. 2019;8(9):1013. doi: 10.3390/cells8091013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Kalra R.S., Bapat S.A. Enhanced levels of double-strand DNA break repair proteins protect ovarian cancer cells against genotoxic stress-induced apoptosis. J Ovarian Res. 2013;6(1):66. doi: 10.1186/1757-2215-6-66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Caccuri F., Sommariva M., Marsico S., et al. Inhibition of DNA repair mechanisms and induction of apoptosis in triple negative breast cancer cells expressing the human herpesvirus 6 U94. Cancers. 2019;11(7):1006. doi: 10.3390/cancers11071006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Kalra R.S., Bapat S.A. Expression proteomics predicts loss of RXR-γ during progression of epithelial ovarian cancer. PLoS One. 2013;8(8):e70398. doi: 10.1371/journal.pone.0070398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Huang C., Vaishnavi K., Kalra R. 3β-methoxy derivation of withaferin-a attenuates its anticancer potency: bioinformatics and molecular evidences. Med Aromatic Plants. 2015;4(219) [Google Scholar]
  • 14.Chaudhary A., Kalra R.S., Malik V., et al. 2, 3-dihydro-3β-methoxy withaferin-A lacks anti-metastasis potency: bioinformatics and experimental evidences. Sci Rep. 2019;9(1):17344. doi: 10.1038/s41598-019-53568-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Chen Y., Jia Y., Song W., Zhang L. Therapeutic potential of nitrogen mustard based hybrid molecules. Front Pharmacol. 2018;9:1453. doi: 10.3389/fphar.2018.01453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Needham D.M., Cohen J.A., Barrett A.M. The mechanism of damage to the bone marrow in systemic poisoning with mustard gas. Biochem J. 1947;41(4):631–639. doi: 10.1042/bj0410631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Ghanei M. Delayed haematological complications of mustard gas. J Appl Toxicol. 2004;24(6):493–495. doi: 10.1002/jat.1006. [DOI] [PubMed] [Google Scholar]
  • 18.Cairns J. The treatment of diseases and the war against cancer. Sci Am. 1985;253(5):51–59. doi: 10.1038/scientificamerican1185-51. [DOI] [PubMed] [Google Scholar]
  • 19.Duffy M.J. The war on cancer: are we winning? Tumour Biol. 2013;34(3):1275–1284. doi: 10.1007/s13277-013-0759-2. [DOI] [PubMed] [Google Scholar]
  • 20.Christakis P. The birth of chemotherapy at Yale. Bicentennial lecture series: surgery Grand Round. Yale J Biol Med. 2011;84(2):169–172. [PMC free article] [PubMed] [Google Scholar]
  • 21.DeVita V.T., Jr., Chu E. A history of cancer chemotherapy. Cancer Res. 2008;68(21):8643–8653. doi: 10.1158/0008-5472.CAN-07-6611. [DOI] [PubMed] [Google Scholar]
  • 22.Sawyers C. Targeted cancer therapy. Nature. 2004;432(7015):294–297. doi: 10.1038/nature03095. [DOI] [PubMed] [Google Scholar]
  • 23.Dewanjee S., Vallamkondu J., Kalra R.S., et al. The emerging role of HDACs: pathology and therapeutic targets in diabetes mellitus. Cells. 2021;10(6):1340. doi: 10.3390/cells10061340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Mattheolabakis G., Rigas B., Constantinides P.P. Nanodelivery strategies in cancer chemotherapy: biological rationale and pharmaceutical perspectives. Nanomedicine. 2012;7(10):1577–1590. doi: 10.2217/nnm.12.128. [DOI] [PubMed] [Google Scholar]
  • 25.Leu J.I.J., Pimkina J., Pandey P., Murphy M.E., George D.L. HSP70 inhibition by the small-molecule 2-phenylethynesulfonamide impairs protein clearance pathways in tumor cells. Mol Cancer Res. 2011;9(7):936–947. doi: 10.1158/1541-7786.MCR-11-0019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Wu W.K., Wu Y.C., Yu L., Li Z.J., Sung J.J., Cho C.H. Induction of autophagy by proteasome inhibitor is associated with proliferative arrest in colon cancer cells. Biochem Biophys Res Commun. 2008;374(2):258–263. doi: 10.1016/j.bbrc.2008.07.031. [DOI] [PubMed] [Google Scholar]
  • 27.Selimovic D., Porzig B.B.O.W., El-Khattouti A., et al. Bortezomib/proteasome inhibitor triggers both apoptosis and autophagy-dependent pathways in melanoma cells. Cell Signal. 2013;25(1):308–318. doi: 10.1016/j.cellsig.2012.10.004. [DOI] [PubMed] [Google Scholar]
  • 28.Hasima N., Ozpolat B. Regulation of autophagy by polyphenolic compounds as a potential therapeutic strategy for cancer. Cell Death Dis. 2014;5:e1509. doi: 10.1038/cddis.2014.467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Sannino S., Guerriero C.J., Sabnis A.J., et al. Compensatory increases of select proteostasis networks after Hsp70 inhibition in cancer cells. J Cell Sci. 2018;131(17):jcs217760. doi: 10.1242/jcs.217760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Zhu K., Dunner K., Jr., McConkey D.J. Proteasome inhibitors activate autophagy as a cytoprotective response in human prostate cancer cells. Oncogene. 2010;29(3):451–462. doi: 10.1038/onc.2009.343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Chen J., Chen X., Xu D., et al. Autophagy induced by proteasomal DUB inhibitor NiPT restricts NiPT-mediated cancer cell death. Front Oncol. 2020;10:348. doi: 10.3389/fonc.2020.00348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Stewart D.A., Winnike J.H., McRitchie S.L., Clark R.F., Pathmasiri W.W., Sumner S.J. Metabolomics analysis of hormone-responsive and triple-negative breast cancer cell responses to paclitaxel identify key metabolic differences. J Proteome Res. 2016;15(9):3225–3240. doi: 10.1021/acs.jproteome.6b00430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Sun H., Zhang A.H., Liu S.B., et al. Cell metabolomics identify regulatory pathways and targets of magnoline against prostate cancer. J Chromatogr B Analyt Technol Biomed Life Sci. 2018;1102–1103:143–151. doi: 10.1016/j.jchromb.2018.10.017. [DOI] [PubMed] [Google Scholar]
  • 34.Kroemer G., Zitvogel L. Immune checkpoint inhibitors. J Exp Med. 2021;218(3) doi: 10.1084/jem.20201979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Torino F., Barnabei A., Paragliola R., Baldelli R., Appetecchia M., Corsello S.M. Thyroid dysfunction as an unintended side effect of anticancer drugs. Thyroid. 2013;23(11):1345–1366. doi: 10.1089/thy.2013.0241. [DOI] [PubMed] [Google Scholar]
  • 36.Mahner S., Eulenburg C., Staehle A., et al. Prognostic impact of the time interval between surgery and chemotherapy in advanced ovarian cancer: analysis of prospective randomised phase III trials. Eur J Cancer. 2013;49(1):142–149. doi: 10.1016/j.ejca.2012.07.023. [DOI] [PubMed] [Google Scholar]
  • 37.di Lonardo A., Nasi S., Pulciani S. Cancer: we should not forget the past. J Cancer. 2015;6(1):29–39. doi: 10.7150/jca.10336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Papac R.J. Origins of cancer therapy. Yale J Biol Med. 2001;74(6):391–398. [PMC free article] [PubMed] [Google Scholar]
  • 39.Morrison W.B. Cancer chemotherapy: an annotated history. J Vet Intern Med. 2010;24(6):1249–1262. doi: 10.1111/j.1939-1676.2010.0590.x. [DOI] [PubMed] [Google Scholar]
  • 40.Riddle J.M. Ancient and medieval chemotherapy for cancer. Isis. 1985;76(3):319–330. doi: 10.1086/353876. [DOI] [PubMed] [Google Scholar]
  • 41.Lee C.T., Huang Y.W., Yang C.H., Huang K.S. Drug delivery systems and combination therapy by using Vinca alkaloids. Curr Top Med Chem. 2015;15(15):1491–1500. doi: 10.2174/1568026615666150414120547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Lau Y.C., Lip G.Y.H. New advances in the treatment of atrial fibrillation: focus on stroke prevention. Expet Opin Pharmacother. 2014;15(15):2193–2204. doi: 10.1517/14656566.2014.948418. [DOI] [PubMed] [Google Scholar]
  • 43.DeVita V.T., Hellman S., Rosenberg S.A. 11th ed. Lippincott Williams & Wilkins; Philadelphia, PA: 2018. Cancer, Principles & Practice of Oncology. [Google Scholar]
  • 44.Hanahan D., Weinberg R.A. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
  • 45.Caon I., Bartolini B., Parnigoni A., et al. Revisiting the hallmarks of cancer: the role of hyaluronan. Semin Cancer Biol. 2020;62:9–19. doi: 10.1016/j.semcancer.2019.07.007. [DOI] [PubMed] [Google Scholar]
  • 46.Dieci M.V., Arnedos M., Andre F., Soria J.C. Fibroblast growth factor receptor inhibitors as a cancer treatment: from a biologic rationale to medical perspectives. Cancer Discov. 2013;3(3):264–279. doi: 10.1158/2159-8290.CD-12-0362. [DOI] [PubMed] [Google Scholar]
  • 47.Holderfield M., Deuker M.M., McCormick F., McMahon M. Targeting RAF kinases for cancer therapy: BRAF-mutated melanoma and beyond. Nat Rev Cancer. 2014;14(7):455–467. doi: 10.1038/nrc3760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Kalra R.S., Chaudhary A., Yoon A.R., et al. CARF enrichment promotes epithelial-mesenchymal transition via Wnt/β-catenin signaling: its clinical relevance and potential as a therapeutic target. Oncogenesis. 2018;7(5):39. doi: 10.1038/s41389-018-0048-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Chua T.C., Yan T.D., Saxena A., Morris D.L. Should the treatment of peritoneal carcinomatosis by cytoreductive surgery and hyperthermic intraperitoneal chemotherapy still be regarded as a highly morbid procedure? : a systematic review of morbidity and mortality. Ann Surg. 2009;249(6):900–907. doi: 10.1097/SLA.0b013e3181a45d86. [DOI] [PubMed] [Google Scholar]
  • 50.Jin J.F., Zhu L.L., Chen M., et al. The optimal choice of medication administration route regarding intravenous, intramuscular, and subcutaneous injection. Patient Prefer Adherence. 2015;9:923–942. doi: 10.2147/PPA.S87271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Giger-Pabst U., Bucur P., Roger S., et al. Comparison of tissue and blood concentrations of oxaliplatin administrated by different modalities of intraperitoneal chemotherapy. Ann Surg Oncol. 2019;26(13):4445–4451. doi: 10.1245/s10434-019-07695-z. [DOI] [PubMed] [Google Scholar]
  • 52.Lichota A., Gwozdzinski K. Anticancer activity of natural compounds from plant and marine environment. Int J Mol Sci. 2018;19(11):3533. doi: 10.3390/ijms19113533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Weaver B.A. How Taxol/paclitaxel kills cancer cells. Mol Biol Cell. 2014;25(18):2677–2681. doi: 10.1091/mbc.E14-04-0916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Omar A., Kalra R.S., Putri J., Elwakeel A., Kaul S.C., Wadhwa R. Soyasapogenol-A targets CARF and results in suppression of tumor growth and metastasis in p53 compromised cancer cells. Sci Rep. 2020;10(1):6323. doi: 10.1038/s41598-020-62953-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Sun G., Rong D., Li Z., et al. Role of small molecule targeted compounds in cancer: progress, opportunities, and challenges. Front Cell Dev Biol. 2021;9:694363. doi: 10.3389/fcell.2021.694363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Apetoh L., Ladoire S., Coukos G., Ghiringhelli F. Combining immunotherapy and anticancer agents: the right path to achieve cancer cure? Ann Oncol. 2015;26(9):1813–1823. doi: 10.1093/annonc/mdv209. [DOI] [PubMed] [Google Scholar]
  • 57.Piktel E., Niemirowicz K., Wątek M., Wollny T., Deptuła P., Bucki R. Recent insights in nanotechnology-based drugs and formulations designed for effective anti-cancer therapy. J Nanobiotechnol. 2016;14(1):39. doi: 10.1186/s12951-016-0193-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Anand U., Carpena M., Kowalska-Góralska M., et al. Safer plant-based nanoparticles for combating antibiotic resistance in bacteria: a comprehensive review on its potential applications, recent advances, and future perspective. Sci Total Environ. 2022:153472. doi: 10.1016/j.scitotenv.2022.153472. [DOI] [PubMed] [Google Scholar]
  • 59.Kaur K., Reddy S., Barathe P., et al. Combating drug-resistant bacteria using photothermally active nanomaterials: a perspective review. Front Microbiol. 2021;12:747019. doi: 10.3389/fmicb.2021.747019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Mitra A.K., Agrahari V., Mandal A., et al. Novel delivery approaches for cancer therapeutics. J Contr Release. 2015;219:248–268. doi: 10.1016/j.jconrel.2015.09.067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Large D.E., Soucy J.R., Hebert J., Auguste D.T. Advances in receptor-mediated, tumor-targeted drug delivery. Adv Ther. 2019;2(1):1800091. doi: 10.1002/adtp.201800091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Yarchoan M., Agarwal P., Villanueva A., et al. Recent developments and therapeutic strategies against hepatocellular carcinoma. Cancer Res. 2019;79(17):4326–4330. doi: 10.1158/0008-5472.CAN-19-0803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Vaidyanathan R., Soon R.H., Zhang P., Jiang K., Lim C.T. Cancer diagnosis: from tumor to liquid biopsy and beyond. Lab Chip. 2018;19(1):11–34. doi: 10.1039/c8lc00684a. [DOI] [PubMed] [Google Scholar]
  • 64.Bera K., Schalper K.A., Rimm D.L., Velcheti V., Madabhushi A. Artificial intelligence in digital pathology - new tools for diagnosis and precision oncology. Nat Rev Clin Oncol. 2019;16(11):703–715. doi: 10.1038/s41571-019-0252-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Charmsaz S., Prencipe M., Kiely M., Pidgeon G.P., Collins D.M. Innovative technologies changing cancer treatment. Cancers. 2018;10(6):208. doi: 10.3390/cancers10060208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Markham M.J., Wachter K., Agarwal N., et al. Clinical cancer advances 2020:annual report on progress against cancer from the American society of clinical oncology. J Clin Oncol. 2020;38(10):1081. doi: 10.1200/JCO.19.03141. [DOI] [PubMed] [Google Scholar]
  • 67.Jaffee E.M., van Dang C., Agus D.B., et al. Future cancer research priorities in the USA: a lancet oncology commission. Lancet Oncol. 2017;18(11):e653–e706. doi: 10.1016/S1470-2045(17)30698-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Zhao J., Dean D.C., Hornicek F.J., Yu X., Duan Z. Emerging next-generation sequencing-based discoveries for targeted osteosarcoma therapy. Cancer Lett. 2020;474:158–167. doi: 10.1016/j.canlet.2020.01.020. [DOI] [PubMed] [Google Scholar]
  • 69.Coccia M. Deep learning technology for improving cancer care in society: new directions in cancer imaging driven by artificial intelligence. Technol Soc. 2020;60:101198. [Google Scholar]
  • 70.Patel S. Emerging adjuvant therapy for cancer: propolis and its constituents. J Diet Suppl. 2016;13(3):245–268. doi: 10.3109/19390211.2015.1008614. [DOI] [PubMed] [Google Scholar]
  • 71.Grob J.J., Garbe C., Ascierto P., Larkin J., Dummer R., Schadendorf D. Adjuvant melanoma therapy with new drugs: should physicians continue to focus on metastatic disease or use it earlier in primary melanoma? Lancet Oncol. 2018;19(12):e720–e725. doi: 10.1016/S1470-2045(18)30596-5. [DOI] [PubMed] [Google Scholar]
  • 72.Benson A.B., 3rd, Schrag D., Somerfield M.R., et al. American Society of Clinical Oncology recommendations on adjuvant chemotherapy for stage II colon cancer. J Clin Oncol. 2004;22(16):3408–3419. doi: 10.1200/JCO.2004.05.063. [DOI] [PubMed] [Google Scholar]
  • 73.Matei D., Filiaci V., Randall M.E., et al. Adjuvant chemotherapy plus radiation for locally advanced endometrial cancer. N Engl J Med. 2019;380(24):2317–2326. doi: 10.1056/NEJMoa1813181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Cao L., Xu C., Cai G., et al. How does the interval between completion of adjuvant chemotherapy and initiation of radiotherapy impact clinical outcomes in operable breast cancer patients? Ann Surg Oncol. 2021;28(4):2155–2168. doi: 10.1245/s10434-020-09026-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Salazar M.C., Rosen J.E., Wang Z., et al. Association of delayed adjuvant chemotherapy with survival after lung cancer surgery. JAMA Oncol. 2017;3(5):610–619. doi: 10.1001/jamaoncol.2016.5829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Kalyanaraman B. Teaching the basics of cancer metabolism: developing antitumor strategies by exploiting the differences between normal and cancer cell metabolism. Redox Biol. 2017;12:833–842. doi: 10.1016/j.redox.2017.04.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Moore A. Metabolic cycles in cancer cells? Bioessays. 2020;42(4):e2000048. doi: 10.1002/bies.202000048. [DOI] [PubMed] [Google Scholar]
  • 78.Anand U., Jacobo-Herrera N., Altemimi A., Lakhssassi N. A comprehensive review on medicinal plants as antimicrobial therapeutics: potential avenues of biocompatible drug discovery. Metabolites. 2019;9(11):258. doi: 10.3390/metabo9110258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Alibek K., Bekmurzayeva A., Mussabekova A., Sultankulov B. Using antimicrobial adjuvant therapy in cancer treatment: a review. Infect Agents Cancer. 2012;7(1):33. doi: 10.1186/1750-9378-7-33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Shoshan-Barmatz V., Anand U., Nahon-Crystal E., di Carlo M., Shteinfer-Kuzmine A. Adverse effects of metformin from diabetes to COVID-19, cancer, neurodegenerative diseases, and aging: is VDAC1 a common target? Front Physiol. 2021;12:730048. doi: 10.3389/fphys.2021.730048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Nicholson L.B. The immune system. Essays Biochem. 2016;60(3):275–301. doi: 10.1042/EBC20160017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Gonzalez H., Hagerling C., Werb Z. Roles of the immune system in cancer: from tumor initiation to metastatic progression. Genes Dev. 2018;32(19–20):1267–1284. doi: 10.1101/gad.314617.118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Whiteside T.L. Immune responses to cancer: are they potential biomarkers of prognosis? Front Oncol. 2013;3:107. doi: 10.3389/fonc.2013.00107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Kreuzaler P., Panina Y., Segal J., Yuneva M. Adapt and conquer: metabolic flexibility in cancer growth, invasion and evasion. Mol Metabol. 2020;33:83–101. doi: 10.1016/j.molmet.2019.08.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Yoshida G.J. Metabolic reprogramming: the emerging concept and associated therapeutic strategies. J Exp Clin Cancer Res. 2015;34:111. doi: 10.1186/s13046-015-0221-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Wadhwa R., Kalra R.S., Kaul S.C. CARF is a multi-module regulator of cell proliferation and a molecular bridge between cellular senescence and carcinogenesis. Mech Ageing Dev. 2017;166:64–68. doi: 10.1016/j.mad.2017.07.008. [DOI] [PubMed] [Google Scholar]
  • 87.Mroz E.A., Rocco J.W. The challenges of tumor genetic diversity. Cancer. 2017;123(6):917–927. doi: 10.1002/cncr.30430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Gambara G., Gaebler M., Keilholz U., Regenbrecht C.R.A., Silvestri A. From chemotherapy to combined targeted therapeutics: in vitro and in vivo models to decipher intra-tumor heterogeneity. Front Pharmacol. 2018;9:77. doi: 10.3389/fphar.2018.00077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Palmer A.C., Sorger P.K. Combination cancer therapy can confer benefit via patient-to-patient variability without drug additivity or synergy. Cell. 2017;171(7):1678–1691. doi: 10.1016/j.cell.2017.11.009. e13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Ke X., Shen L. Molecular targeted therapy of cancer: the progress and future prospect. Front Lab Med. 2017;1(2):69–75. [Google Scholar]
  • 91.Kondo N., Takahashi A., Ono K., Ohnishi T. DNA damage induced by alkylating agents and repair pathways. J Nucleic Acids. 2010;2010:543531. doi: 10.4061/2010/543531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Qiu H., Wang Y. Exploring DNA-binding proteins with in vivo chemical cross-linking and mass spectrometry. J Proteome Res. 2009;8(4):1983–1991. doi: 10.1021/pr8009319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Mehrmohamadi M., Jeong S.H., Locasale J.W. Molecular features that predict the response to antimetabolite chemotherapies. Cancer Metabol. 2017;5:8. doi: 10.1186/s40170-017-0170-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Katzung B.G. 10th ed. McGraw-Hill; Singapore: 2008. Basic and Clinical Pharmacology. [Google Scholar]
  • 95.Lambie D.G., Johnson R.H. Drugs and folate metabolism. Drugs. 1985;30(2):145–155. doi: 10.2165/00003495-198530020-00003. [DOI] [PubMed] [Google Scholar]
  • 96.Raimondi M.V., Randazzo O., la Franca M., et al. DHFR inhibitors: reading the past for discovering novel anticancer agents. Molecules. 2019;24(6):1140. doi: 10.3390/molecules24061140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Löwenberg B., Pabst T., Vellenga E., et al. Cytarabine dose for acute myeloid leukemia. N Engl J Med. 2011;364(11):1027–1036. doi: 10.1056/NEJMoa1010222. [DOI] [PubMed] [Google Scholar]
  • 98.Grem J.L. 5-Fluorouracil: forty-plus and still ticking. A review of its preclinical and clinical development. Invest N Drugs. 2000;18(4):299–313. doi: 10.1023/a:1006416410198. [DOI] [PubMed] [Google Scholar]
  • 99.Masuda N., Lee S.J., Ohtani S., et al. Adjuvant capecitabine for breast cancer after preoperative chemotherapy. N Engl J Med. 2017;376(22):2147–2159. doi: 10.1056/NEJMoa1612645. [DOI] [PubMed] [Google Scholar]
  • 100.Dyawanapelly S., Kumar A., Chourasia M.K. Lessons learned from gemcitabine: impact of therapeutic carrier systems and gemcitabine's drug conjugates on cancer therapy. Crit Rev Ther Drug Carrier Syst. 2017;34(1):63–96. doi: 10.1615/CritRevTherDrugCarrierSyst.2017017912. [DOI] [PubMed] [Google Scholar]
  • 101.Malik P., Cashen A.F. Decitabine in the treatment of acute myeloid leukemia in elderly patients. Cancer Manag Res. 2014;6:53–61. doi: 10.2147/CMAR.S40600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Liu Y., Wu W., Hong W., Sun X., Wu J., Huang Q. Raltitrexed-based chemotherapy for advanced colorectal cancer. Clin Res Hepatol Gastroenterol. 2014;38(2):219–225. doi: 10.1016/j.clinre.2013.11.006. [DOI] [PubMed] [Google Scholar]
  • 103.Hasegawa K., Saiura A., Takayama T., et al. Oral adjuvant chemotherapy using uracil-tegafur (UFT) with leucovorin (LV) after resection of colorectal cancer liver metastases: long-term survival results of the phase III UFT/LV study. J Clin Oncol. 2017;35:672. [Google Scholar]
  • 104.Moudi M., Go R., Yien C.Y., Nazre M. Vinca alkaloids. Int J Prev Med. 2013;4(11):1231–1235. [PMC free article] [PubMed] [Google Scholar]
  • 105.Bates D., Eastman A. Microtubule destabilising agents: far more than just antimitotic anticancer drugs. Br J Clin Pharmacol. 2017;83(2):255–268. doi: 10.1111/bcp.13126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Habli Z., Toumieh G., Fatfat M., Rahal O.N., Gali-Muhtasib H. Emerging cytotoxic alkaloids in the battle against cancer: overview of molecular mechanisms. Molecules. 2017;22(2):250. doi: 10.3390/molecules22020250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Falzone L., Salomone S., Libra M. Evolution of cancer pharmacological treatments at the turn of the third millennium. Front Pharmacol. 2018;9:1300. doi: 10.3389/fphar.2018.01300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Schneider F., Pan L., Ottenbruch M., List T., Gaich T. The chemistry of nonclassical taxane diterpene. Acc Chem Res. 2021;54(10):2347–2360. doi: 10.1021/acs.accounts.0c00873. [DOI] [PubMed] [Google Scholar]
  • 109.Fanale D., Bronte G., Passiglia F., et al. Stabilizing versus destabilizing the microtubules: a double-edge sword for an effective cancer treatment option? Anal Cell Pathol. 2015;2015:690916. doi: 10.1155/2015/690916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Leung J.C., Cassimeris L. Reorganization of paclitaxel-stabilized microtubule arrays at mitotic entry: roles of depolymerizing kinesins and severing proteins. Cancer Biol Ther. 2019;20(10):1337–1347. doi: 10.1080/15384047.2019.1638678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Schwab C.L., English D.P., Roque D.M., Santin A.D. Taxanes: their impact on gynecologic malignancy. Anti Cancer Drugs. 2014;25(5):522–535. doi: 10.1097/CAD.0000000000000057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Abu Samaan T.M., Samec M., Liskova A., Kubatka P., Büsselberg D. Paclitaxel's mechanistic and clinical effects on breast cancer. Biomolecules. 2019;9(12):789. doi: 10.3390/biom9120789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Zhu L., Chen L. Progress in research on paclitaxel and tumor immunotherapy. Cell Mol Biol Lett. 2019;24:40. doi: 10.1186/s11658-019-0164-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Mahalaxmi I., Devi S.M., Kaavya J., Arul N., Balachandar V., Santhy K.S. New insight into NANOG: a novel therapeutic target for ovarian cancer (OC) Eur J Pharmacol. 2019;852:51–57. doi: 10.1016/j.ejphar.2019.03.003. [DOI] [PubMed] [Google Scholar]
  • 115.Postma T.J., Vermorken J.B., Liefting A.J., Pinedo H.M., Heimans J.J. Paclitaxel-induced neuropathy. Ann Oncol. 1995;6(5):489–494. doi: 10.1093/oxfordjournals.annonc.a059220. [DOI] [PubMed] [Google Scholar]
  • 116.Das T., Anand U., Pandey S.K., et al. Therapeutic strategies to overcome taxane resistance in cancer. Drug Resist Updates. 2021;55:100754. doi: 10.1016/j.drup.2021.100754. [DOI] [PubMed] [Google Scholar]
  • 117.Stähelin H.F., von Wartburg A. The chemical and biological route from podophyllotoxin glucoside to etoposide: ninth Cain memorial Award lecture. Cancer Res. 1991;51(1):5–15. [PubMed] [Google Scholar]
  • 118.Green J.A., Tarpey A.W., Warenius H.M. Pharmacokinetic study of high dose etoposide infusion in patients with small cell lung cancer. Acta Oncol. 1988;27(6b):819–822. doi: 10.3109/02841868809094365. [DOI] [PubMed] [Google Scholar]
  • 119.Infante Lara L., Fenner S., Ratcliffe S., et al. Coupling the core of the anticancer drug etoposide to an oligonucleotide induces topoisomerase II-mediated cleavage at specific DNA sequences. Nucleic Acids Res. 2018;46(5):2218–2233. doi: 10.1093/nar/gky072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Pommier Y., Leo E., Zhang H., Marchand C. DNA topoisomerases and their poisoning by anticancer and antibacterial drugs. Chem Biol. 2010;17(5):421–433. doi: 10.1016/j.chembiol.2010.04.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Bruni E., Reichle A., Scimeca M., Bonanno E., Ghibelli L. Lowering etoposide doses shifts cell demise from caspase-dependent to differentiation and caspase-3-independent apoptosis via DNA damage response, inducing AML culture extinction. Front Pharmacol. 2018;9:1307. doi: 10.3389/fphar.2018.01307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Hande K.R. Etoposide: four decades of development of a topoisomerase II inhibitor. Eur J Cancer. 1998;34(10):1514–1521. doi: 10.1016/s0959-8049(98)00228-7. [DOI] [PubMed] [Google Scholar]
  • 123.Wall M.E. Camptothecin and taxol: discovery to clinic. Med Res Rev. 1998;18(5):299–314. doi: 10.1002/(sici)1098-1128(199809)18:5<299::aid-med2>3.0.co;2-o. [DOI] [PubMed] [Google Scholar]
  • 124.Martino E., Della Volpe S., Terribile E., et al. The long story of camptothecin: from traditional medicine to drugs. Bioorg Med Chem Lett. 2017;27(4):701–707. doi: 10.1016/j.bmcl.2016.12.085. [DOI] [PubMed] [Google Scholar]
  • 125.Yu S., Huang Q.Q., Luo Y., Lu W. Total synthesis of camptothecin and SN-38. J Org Chem. 2012;77(1):713–717. doi: 10.1021/jo201974f. [DOI] [PubMed] [Google Scholar]
  • 126.Li F., Jiang T., Li Q., Ling X. Camptothecin (CPT) and its derivatives are known to target topoisomerase I (Top1) as their mechanism of action: did we miss something in CPT analogue molecular targets for treating human disease such as cancer? Am J Cancer Res. 2017;7(12):2350–2394. [PMC free article] [PubMed] [Google Scholar]
  • 127.Guo Y., Shi M., Shen X., Yang C., Yang L., Zhang J. Capecitabine plus irinotecan versus 5-FU/leucovorin plus irinotecan in the treatment of colorectal cancer: a meta-analysis. Clin Colorectal Cancer. 2014;13(2):110–118. doi: 10.1016/j.clcc.2013.12.004. [DOI] [PubMed] [Google Scholar]
  • 128.Herben V.M., ten Bokkel Huinink W.W., Beijnen J.H. Clinical pharmacokinetics of topotecan. Clin Pharmacokinet. 1996;31(2):85–102. doi: 10.2165/00003088-199631020-00001. [DOI] [PubMed] [Google Scholar]
  • 129.Pommier Y. Topoisomerase I inhibitors: camptothecins and beyond. Nat Rev Cancer. 2006;6(10):789–802. doi: 10.1038/nrc1977. [DOI] [PubMed] [Google Scholar]
  • 130.Tomicic M.T., Christmann M., Kaina B. Topotecan-triggered degradation of topoisomerase I is p53-dependent and impacts cell survival. Cancer Res. 2005;65(19):8920–8926. doi: 10.1158/0008-5472.CAN-05-0266. [DOI] [PubMed] [Google Scholar]
  • 131.Bailly C. Irinotecan: 25 years of cancer treatment. Pharmacol Res. 2019;148:104398. doi: 10.1016/j.phrs.2019.104398. [DOI] [PubMed] [Google Scholar]
  • 132.Sawada S., Yokokura T., Miyasaka T. Synthesis of CPT-11 (irinotecan hydrochloride trihydrate) Ann N Y Acad Sci. 1996;803:13–28. doi: 10.1111/j.1749-6632.1996.tb26372.x. [DOI] [PubMed] [Google Scholar]
  • 133.Sobell H.M. Actinomycin and DNA transcription. Proc Natl Acad Sci USA. 1985;82(16):5328–5331. doi: 10.1073/pnas.82.16.5328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Avendaño C., Menéndez J.C. Elsevier; Amsterdam: 2008. Medicinal Chemistry of Anticancer Drugs. [Google Scholar]
  • 135.Arndt C., Hawkins D., Anderson J.R., Breitfeld P., Womer R., Meyer W. Age is a risk factor for chemotherapy-induced hepatopathy with vincristine, dactinomycin, and cyclophosphamide. J Clin Oncol. 2004;22(10):1894–1901. doi: 10.1200/JCO.2004.08.075. [DOI] [PubMed] [Google Scholar]
  • 136.Al-Hazmi A., Zwaan J., Awad A., Al-Mesfer S., Mullaney P.B., Wheeler D.T. Effectiveness and complications of mitomycin C use during pediatric glaucoma surgery. Ophthalmology. 1998;105(10):1915–1920. doi: 10.1016/S0161-6420(98)91041-7. [DOI] [PubMed] [Google Scholar]
  • 137.Crooke S.T., Bradner W.T. Mitomycin C: a review. Cancer Treat Rev. 1976;3(3):121–139. doi: 10.1016/s0305-7372(76)80019-9. [DOI] [PubMed] [Google Scholar]
  • 138.Tomasz M. Mitomycin C: small, fast and deadly (but very selective) Chem Biol. 1995;2(9):575–579. doi: 10.1016/1074-5521(95)90120-5. [DOI] [PubMed] [Google Scholar]
  • 139.Noll D.M., Mason T.M., Miller P.S. Formation and repair of interstrand cross-links in DNA. Chem Rev. 2006;106(2):277–301. doi: 10.1021/cr040478b. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Haffty B.G., Wilson L.D., Son Y.H., et al. Concurrent chemo-radiotherapy with mitomycin C compared with porfiromycin in squamous cell cancer of the head and neck: final results of a randomized clinical trial. Int J Radiat Oncol Biol Phys. 2005;61(1):119–128. doi: 10.1016/j.ijrobp.2004.07.730. [DOI] [PubMed] [Google Scholar]
  • 141.Murphy T., Yee K.W.L. Cytarabine and daunorubicin for the treatment of acute myeloid leukemia. Expet Opin Pharmacother. 2017;18(16):1765–1780. doi: 10.1080/14656566.2017.1391216. [DOI] [PubMed] [Google Scholar]
  • 142.Tan C., Tasaka H., Yu K.P., Murphy M.L., Karnofsky D.A. Daunomycin, an antitumor antibiotic, in the treatment of neoplastic disease. Clinical evaluation with special reference to childhood leukemia. Cancer. 1967;20(3):333–353. doi: 10.1002/1097-0142(1967)20:3<333::aid-cncr2820200302>3.0.co;2-k. [DOI] [PubMed] [Google Scholar]
  • 143.Fornari F.A., Randolph J.K., Yalowich J.C., Ritke M.K., Gewirtz D.A. Interference by doxorubicin with DNA unwinding in MCF-7 breast tumor cells. Mol Pharmacol. 1994;45(4):649–656. [PubMed] [Google Scholar]
  • 144.Bodley A., Liu L.F., Israel M., et al. DNA topoisomerase II-mediated interaction of doxorubicin and daunorubicin congeners with DNA. Cancer Res. 1989;49(21):5969–5978. [PubMed] [Google Scholar]
  • 145.Zhang G., Fang L., Zhu L., et al. Syntheses and biological activities of disaccharide daunorubicins. J Med Chem. 2005;48(16):5269–5278. doi: 10.1021/jm050144u. [DOI] [PubMed] [Google Scholar]
  • 146.Yu Z., Yan B., Gao L., et al. Targeted delivery of bleomycin: a comprehensive anticancer review. Curr Cancer Drug Targets. 2016;16(6):509–521. doi: 10.2174/1568009616666151130213910. [DOI] [PubMed] [Google Scholar]
  • 147.Lown J.W., Sim S.K. The mechanism of the bleomycin-induced cleavage of DNA. Biochem Biophys Res Commun. 1977;77(4):1150–1157. doi: 10.1016/s0006-291x(77)80099-5. [DOI] [PubMed] [Google Scholar]
  • 148.Weber G.F. Molecular Therapies of Cancer. Springer; Cham.: 2015. DNA damaging drugs. [Google Scholar]
  • 149.Meldrum C., Doyle M.A., Tothill R.W. Next-generation sequencing for cancer diagnostics: a practical perspective. Clin Biochem Rev. 2011;32(4):177–195. [PMC free article] [PubMed] [Google Scholar]
  • 150.Kalra R.S., Cheung C.T., Chaudhary A., Prakash J., Kaul S.C., Wadhwa R. CARF (Collaborator of ARF) overexpression in p53-deficient cells promotes carcinogenesis. Mol Oncol. 2015;9(9):1877–1889. doi: 10.1016/j.molonc.2015.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Joshi V., Upadhyay A., Prajapati V.K., Mishra A. How autophagy can restore proteostasis defects in multiple diseases? Med Res Rev. 2020;40(4):1385–1439. doi: 10.1002/med.21662. [DOI] [PubMed] [Google Scholar]
  • 152.Mishra R., Upadhyay A., Prajapati V.K., Mishra A. Proteasome-mediated proteostasis: novel medicinal and pharmacological strategies for diseases. Med Res Rev. 2018;38(6):1916–1973. doi: 10.1002/med.21502. [DOI] [PubMed] [Google Scholar]
  • 153.Upadhyay A., Amanullah A., Chhangani D., Joshi V., Mishra R., Mishra A. Ibuprofen induces mitochondrial-mediated apoptosis through proteasomal dysfunction. Mol Neurobiol. 2016;53(10):6968–6981. doi: 10.1007/s12035-015-9603-6. [DOI] [PubMed] [Google Scholar]
  • 154.Na Y., Kaul S.C., Ryu J., et al. Stress chaperone mortalin contributes to epithelial-mesenchymal transition and cancer metastasis. Cancer Res. 2016;76(9):2754–2765. doi: 10.1158/0008-5472.CAN-15-2704. [DOI] [PubMed] [Google Scholar]
  • 155.Bapat S.A., Krishnan A., Ghanate A.D., Kusumbe A.P., Kalra R.S. Gene expression: protein interaction systems network modeling identifies transformation-associated molecules and pathways in ovarian cancer. Cancer Res. 2010;70(12):4809–4819. doi: 10.1158/0008-5472.CAN-10-0447. [DOI] [PubMed] [Google Scholar]
  • 156.Stringer A.M., Gibson R.J., Logan R.M., et al. Irinotecan-induced mucositis is associated with changes in intestinal mucins. Cancer Chemother Pharmacol. 2009;64(1):123–132. doi: 10.1007/s00280-008-0855-y. [DOI] [PubMed] [Google Scholar]
  • 157.Thorpe D., Stringer A., Butler R. Chemotherapy-induced mucositis: the role of mucin secretion and regulation, and the enteric nervous system. Neurotoxicology. 2013;38:101–105. doi: 10.1016/j.neuro.2013.06.007. [DOI] [PubMed] [Google Scholar]
  • 158.Mitchison T.J. The proliferation rate paradox in antimitotic chemotherapy. Mol Biol Cell. 2012;23(1):1–6. doi: 10.1091/mbc.E10-04-0335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.McQuade R.M., Stojanovska V., Abalo R., Bornstein J.C., Nurgali K. Chemotherapy-induced constipation and diarrhea: pathophysiology, current and emerging treatments. Front Pharmacol. 2016;7:414. doi: 10.3389/fphar.2016.00414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Bower J.E. Cancer-related fatigue: mechanisms, risk factors, and treatments. Nat Rev Clin Oncol. 2014;11(10):597–609. doi: 10.1038/nrclinonc.2014.127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Wang Y., Probin V., Zhou D. Cancer therapy-induced residual bone marrow injury-Mechanisms of induction and implication for therapy. Curr Cancer Ther Rev. 2006;2(3):271–279. doi: 10.2174/157339406777934717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Kuter D.J. Managing thrombocytopenia associated with cancer chemotherapy. Oncology. 2015;29(4):282–294. [PubMed] [Google Scholar]
  • 163.Manikandan R., Kumar S., Dorairajan L.N. Hemorrhagic cystitis: a challenge to the urologist. Indian J Urol. 2010;26(2):159–166. doi: 10.4103/0970-1591.65380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Horie S., Oya M., Nangaku M., et al. Guidelines for treatment of renal injury during cancer chemotherapy 2016. Clin Exp Nephrol. 2018;22(1):210–244. doi: 10.1007/s10157-017-1448-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Maor Y., Malnick S. Liver injury induced by anticancer chemotherapy and radiation therapy. Int J Hepatol. 2013;2013:815105. doi: 10.1155/2013/815105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Yeh E.T., Tong A.T., Lenihan D.J., et al. Cardiovascular complications of cancer therapy: diagnosis, pathogenesis, and management. Circulation. 2004;109(25):3122–3131. doi: 10.1161/01.CIR.0000133187.74800.B9. [DOI] [PubMed] [Google Scholar]
  • 167.Schover L.R., van der Kaaij M., van Dorst E., Creutzberg C., Huyghe E., Kiserud C.E. Sexual dysfunction and infertility as late effects of cancer treatment. EJC Suppl. 2014;12(1):41–53. doi: 10.1016/j.ejcsup.2014.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Voznesensky M., Annam K., Kreder K.J. Understanding and managing erectile dysfunction in patients treated for cancer. J Oncol Pract. 2016;12(4):297–304. doi: 10.1200/JOP.2016.010678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Mitchell T., Turton P. 'Chemobrain': concentration and memory effects in people receiving chemotherapy - a descriptive phenomenological study. Eur J Cancer Care. 2011;20(4):539–548. doi: 10.1111/j.1365-2354.2011.01244.x. [DOI] [PubMed] [Google Scholar]
  • 170.Moore H.C. An overview of chemotherapy-related cognitive dysfunction, or ‘chemobrain. Oncology. 2014;28(9):797–804. [PubMed] [Google Scholar]
  • 171.Dutta V. Chemotherapy, neurotoxicity, and cognitive changes in breast cancer. J Cancer Res Therapeut. 2011;7(3):264–269. doi: 10.4103/0973-1482.87008. [DOI] [PubMed] [Google Scholar]
  • 172.Omoti A., Omoti C. Ocular toxicity of systemic anticancer chemotherapy. Pharm Pract. 2006;4:55–59. [PMC free article] [PubMed] [Google Scholar]
  • 173.Al-Tweigeri T., Nabholtz J.M., MacKey J.R. Ocular toxicity and cancer chemotherapy. A review. Cancer. 1996;78(7):1359–1373. doi: 10.1002/(SICI)1097-0142(19961001)78:7<1359::AID-CNCR1>3.0.CO;2-G. [DOI] [PubMed] [Google Scholar]
  • 174.Curigliano G., Cardinale D., Suter T., et al. Cardiovascular toxicity induced by chemotherapy, targeted agents and radiotherapy: ESMO Clinical Practice Guidelines. Ann Oncol. 2012;23(Suppl 7):vii155–vii166. doi: 10.1093/annonc/mds293. [DOI] [PubMed] [Google Scholar]
  • 175.Cohen J.D., Li L., Wang Y., et al. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science. 2018;359(6378):926–930. doi: 10.1126/science.aar3247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Alix-Panabières C., Pantel K. Clinical applications of circulating tumor cells and circulating tumor DNA as liquid biopsy. Cancer Discov. 2016;6(5):479–491. doi: 10.1158/2159-8290.CD-15-1483. [DOI] [PubMed] [Google Scholar]
  • 177.Li L., Gao R., Yu Y., et al. Tumor suppressor activity of miR-451:identification of CARF as a new target. Sci Rep. 2018;8(1):375. doi: 10.1038/s41598-017-18559-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Shohdy K.S., West H.J. Circulating tumor DNA testing-liquid biopsy of a cancer. JAMA Oncol. 2020;6(5):792. doi: 10.1001/jamaoncol.2020.0346. [DOI] [PubMed] [Google Scholar]
  • 179.Vivek R., Thangam R., NipunBabu V., et al. Multifunctional HER2-antibody conjugated polymeric nanocarrier-based drug delivery system for multi-drug-resistant breast cancer therapy. ACS Appl Mater Interfaces. 2014;6(9):6469–6480. doi: 10.1021/am406012g. [DOI] [PubMed] [Google Scholar]
  • 180.Alley S.C., Okeley N.M., Senter P.D. Antibody-drug conjugates: targeted drug delivery for cancer. Curr Opin Chem Biol. 2010;14(4):529–537. doi: 10.1016/j.cbpa.2010.06.170. [DOI] [PubMed] [Google Scholar]
  • 181.Wang X., Wang Y., Chen Z.G., Shin D.M. Advances of cancer therapy by nanotechnology. Cancer Res Treat. 2009;41(1):1–11. doi: 10.4143/crt.2009.41.1.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Firer M.A., Gellerman G. Targeted drug delivery for cancer therapy: the other side of antibodies. J Hematol Oncol. 2012;5:70. doi: 10.1186/1756-8722-5-70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Kutova O.M., Guryev E.L., Sokolova E.A., Alzeibak R., Balalaeva I.V. Targeted delivery to tumors: multidirectional strategies to improve treatment efficiency. Cancers. 2019;11(1):68. doi: 10.3390/cancers11010068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Cho K., Wang X., Nie S., Chen Z.G., Shin D.M. Therapeutic nanoparticles for drug delivery in cancer. Clin Cancer Res. 2008;14(5):1310–1316. doi: 10.1158/1078-0432.CCR-07-1441. [DOI] [PubMed] [Google Scholar]
  • 185.Rosenblum D., Joshi N., Tao W., Karp J.M., Peer D. Progress and challenges towards targeted delivery of cancer therapeutics. Nat Commun. 2018;9(1):1410. doi: 10.1038/s41467-018-03705-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Sutradhar K.B., Amin M.L. Nanotechnology in cancer drug delivery and selective targeting. ISRN Nanotechnol. 2014;2014:939378. [Google Scholar]
  • 187.Lammers T., Kiessling F., Ashford M., Hennink W., Crommelin D., Storm G. Cancer nanomedicine: is targeting our target? Nat Rev Mater. 2016;1(9):16069. doi: 10.1038/natrevmats.2016.69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.El-Readi M.Z., Althubiti M.A. Cancer nanomedicine: a new era of successful targeted therapy. J Nanomater. 2019;2019:4927312. [Google Scholar]
  • 189.Dias-Santagata D., Akhavanfard S., David S.S., et al. Rapid targeted mutational analysis of human tumours: a clinical platform to guide personalized cancer medicine. EMBO Mol Med. 2010;2(5):146–158. doi: 10.1002/emmm.201000070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Dong L., Wang W., Li A., et al. Clinical next generation sequencing for precision medicine in cancer. Curr Genom. 2015;16(4):253–263. doi: 10.2174/1389202915666150511205313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Fedorenko I.V., Gibney G.T., Sondak V.K., Smalley K.S.M. Beyond BRAF: where next for melanoma therapy? Br J Cancer. 2015;112(2):217–226. doi: 10.1038/bjc.2014.476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Zaman A., Wu W., Bivona T.G. Targeting oncogenic BRAF: past, present, and future. Cancers. 2019;11(8):1197. doi: 10.3390/cancers11081197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.André F., Ciruelos E., Rubovszky G., et al. Alpelisib for PIK3CA-mutated, hormone receptor-positive advanced breast cancer. N Engl J Med. 2019;380(20):1929–1940. doi: 10.1056/NEJMoa1813904. [DOI] [PubMed] [Google Scholar]
  • 194.Kalra R.S., Bapat S.A. Springer; New York: 2019. Proteomics to Predict Loss of RXR-γ during Progression of Epithelial Ovarian Cancer. Methods in Molecular Biology. New York, NY; pp. 1–14. [DOI] [PubMed] [Google Scholar]
  • 195.Soda M., Choi Y.L., Enomoto M., et al. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature. 2007;448(7153):561–566. doi: 10.1038/nature05945. [DOI] [PubMed] [Google Scholar]
  • 196.Hyman D.M., Puzanov I., Subbiah V., et al. Vemurafenib in multiple nonmelanoma cancers with BRAF V600 mutations. N Engl J Med. 2015;373(8):726–736. doi: 10.1056/NEJMoa1502309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Babina I.S., Turner N.C. Advances and challenges in targeting FGFR signalling in cancer. Nat Rev Cancer. 2017;17(5):318–332. doi: 10.1038/nrc.2017.8. [DOI] [PubMed] [Google Scholar]
  • 198.Tutt A., Robson M., Garber J.E., et al. Oral poly(ADP-ribose) polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 mutations and advanced breast cancer: a proof-of-concept trial. Lancet. 2010;376(9737):235–244. doi: 10.1016/S0140-6736(10)60892-6. [DOI] [PubMed] [Google Scholar]
  • 199.Fong P.C., Boss D.S., Yap T.A., et al. Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N Engl J Med. 2009;361(2):123–134. doi: 10.1056/NEJMoa0900212. [DOI] [PubMed] [Google Scholar]
  • 200.Chaudhary A., Kalra R.S., Huang C., Prakash J., Kaul S.C., Wadhwa R. 2, 3-dihydro-3β-methoxy withaferin-A protects normal cells against stress: molecular evidence of its potent cytoprotective activity. J Nat Prod. 2017;80(10):2756–2760. doi: 10.1021/acs.jnatprod.7b00573. [DOI] [PubMed] [Google Scholar]
  • 201.Friedman A.A., Letai A., Fisher D.E., Flaherty K.T. Precision medicine for cancer with next-generation functional diagnostics. Nat Rev Cancer. 2015;15(12):747–756. doi: 10.1038/nrc4015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.de la Fuente-Núñez C., Silva O.N., Lu T.K., Franco O.L. Antimicrobial peptides: role in human disease and potential as immunotherapies. Pharmacol Ther. 2017;178:132–140. doi: 10.1016/j.pharmthera.2017.04.002. [DOI] [PubMed] [Google Scholar]
  • 203.Felício M.R., Silva O.N., Gonçalves S., Santos N.C., Franco O.L. Peptides with dual antimicrobial and anticancer activities. Front Chem. 2017;5:5. doi: 10.3389/fchem.2017.00005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Hilchie A.L., Hoskin D.W., Power Coombs M.R. Anticancer activities of natural and synthetic peptides. Adv Exp Med Biol. 2019;1117:131–147. doi: 10.1007/978-981-13-3588-4_9. [DOI] [PubMed] [Google Scholar]
  • 205.Mwangi J., Hao X., Lai R., Zhang Z.Y. Antimicrobial peptides: new hope in the war against multidrug resistance. Zool Res. 2019;40(6):488–505. doi: 10.24272/j.issn.2095-8137.2019.062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Qin Y., Qin Z.D., Chen J., et al. From antimicrobial to anticancer peptides: the transformation of peptides. Recent Pat Anti-Cancer Drug Discov. 2019;14(1):70–84. doi: 10.2174/1574892814666190119165157. [DOI] [PubMed] [Google Scholar]
  • 207.Lei J., Sun L., Huang S., et al. The antimicrobial peptides and their potential clinical applications. Am J Transl Res. 2019;11(7):3919–3931. [PMC free article] [PubMed] [Google Scholar]
  • 208.Roudi R., Syn N.L., Roudbary M. Antimicrobial peptides as biologic and immunotherapeutic agents against cancer: a comprehensive overview. Front Immunol. 2017;8:1320. doi: 10.3389/fimmu.2017.01320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Tornesello A.L., Borrelli A., Buonaguro L., Buonaguro F.M., Tornesello M.L. Antimicrobial peptides as anticancer agents: functional properties and biological activities. Molecules. 2020;25(12):2850. doi: 10.3390/molecules25122850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Khare T., Anand U., Dey A., et al. Exploring phytochemicals for combating antibiotic resistance in microbial pathogens. Front Pharmacol. 2021;12:720726. doi: 10.3389/fphar.2021.720726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.van Harten R.M., van Woudenbergh E., van Dijk A., Haagsman H.P. Cathelicidins: immunomodulatory antimicrobials. Vaccines. 2018;6(3):63. doi: 10.3390/vaccines6030063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Wang G., Narayana J.L., Mishra B., et al. Springer Singapore; Singapore: 2019. Design of Antimicrobial Peptides: Progress Made with Human Cathelicidin LL-37. Advances in Experimental Medicine and Biology; pp. 215–240. [DOI] [PubMed] [Google Scholar]
  • 213.Fabisiak A., Murawska N., Fichna J. LL-37:cathelicidin-related antimicrobial peptide with pleiotropic activity. Pharmacol Rep. 2016;68(4):802–808. doi: 10.1016/j.pharep.2016.03.015. [DOI] [PubMed] [Google Scholar]
  • 214.Chen X., Ji S., Si J., et al. Human cathelicidin antimicrobial peptide suppresses proliferation, migration and invasion of oral carcinoma HSC-3 cells via a novel mechanism involving caspase-3 mediated apoptosis. Mol Med Rep. 2020;22(6):5243–5250. doi: 10.3892/mmr.2020.11629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Porter R.J., Murray G.I., Alnabulsi A., et al. Colonic epithelial cathelicidin (LL-37) expression intensity is associated with progression of colorectal cancer and presence of CD8+ T cell infiltrate. J Pathol Clin Res. 2021;7(5):495–506. doi: 10.1002/cjp2.222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Wang J., Cheng M., Law I.K.M., Ortiz C., Sun M., Koon H.W. Cathelicidin suppresses colon cancer metastasis via a P2RX7-dependent mechanism. Mol Ther Oncolytics. 2019;12:195–203. doi: 10.1016/j.omto.2019.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Biswaro L.S., da Costa Sousa M.G., Rezende T.M.B., Dias S.C., Franco O.L. Antimicrobial peptides and nanotechnology, recent advances and challenges. Front Microbiol. 2018;9:855. doi: 10.3389/fmicb.2018.00855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Niemirowicz K., Prokop I., Wilczewska A.Z., et al. Magnetic nanoparticles enhance the anticancer activity of cathelicidin LL-37 peptide against colon cancer cells. Int J Nanomed. 2015;10:3843–3853. doi: 10.2147/IJN.S76104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Fan R., Tong A., Li X., et al. Enhanced antitumor effects by docetaxel/LL37-loaded thermosensitive hydrogel nanoparticles in peritoneal carcinomatosis of colorectal cancer. Int J Nanomed. 2015;10:7291–7305. doi: 10.2147/IJN.S89066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Chauhan S., Dhawan D.K., Saini A., Preet S. Antimicrobial peptides against colorectal cancer-a focused review. Pharmacol Res. 2021;167:105529. doi: 10.1016/j.phrs.2021.105529. [DOI] [PubMed] [Google Scholar]
  • 221.Dolkar T., Trinidad C.M., Nelson K.C., et al. Dermatologic toxicity from novel therapy using antimicrobial peptide LL-37 in melanoma: a detailed examination of the clinicopathologic features. J Cutan Pathol. 2018;45(7):539–544. doi: 10.1111/cup.13262. [DOI] [PubMed] [Google Scholar]
  • 222.Divyashree M., Mani M.K., Reddy D., et al. Clinical applications of antimicrobial peptides (AMPs):where do we stand now? Protein Pept Lett. 2020;27(2):120–134. doi: 10.2174/0929866526666190925152957. [DOI] [PubMed] [Google Scholar]
  • 223.Karlitepe A., Ozalp O., Avci C.B. New approaches for cancer immunotherapy. Tumour Biol. 2015;36(6):4075–4078. doi: 10.1007/s13277-015-3491-2. [DOI] [PubMed] [Google Scholar]
  • 224.Weiner L.M., Murray J.C., Shuptrine C.W. Antibody-based immunotherapy of cancer. Cell. 2012;148(6):1081–1084. doi: 10.1016/j.cell.2012.02.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Vigneron N. Human tumor antigens and cancer immunotherapy. BioMed Res Int. 2015;2015:948501. doi: 10.1155/2015/948501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Kim I., Sanchez K., McArthur H.L., Page D. Immunotherapy in triple-negative breast cancer: present and future. Curr Breast Cancer Rep. 2019;11(4):259–271. [Google Scholar]
  • 227.Marra A., Viale G., Curigliano G. Recent advances in triple negative breast cancer: the immunotherapy era. BMC Med. 2019;17(1):90. doi: 10.1186/s12916-019-1326-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Sambi M., Bagheri L., Szewczuk M.R. Current challenges in cancer immunotherapy: multimodal approaches to improve efficacy and patient response rates. JAMA Oncol. 2019;2019:4508794. doi: 10.1155/2019/4508794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Paz-Ares L., Luft A., Vicente D., et al. Pembrolizumab plus chemotherapy for squamous non-small-cell lung cancer. N Engl J Med. 2018;379(21):2040–2051. doi: 10.1056/NEJMoa1810865. [DOI] [PubMed] [Google Scholar]
  • 230.Finn O.J. Vaccines for cancer prevention: a practical and feasible approach to the cancer epidemic. Cancer Immunol Res. 2014;2(8):708–713. doi: 10.1158/2326-6066.CIR-14-0110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Pérez-García J., Soberino J., Racca F., Gion M., Stradella A., Cortés J. Atezolizumab in the treatment of metastatic triple-negative breast cancer. Expet Opin Biol Ther. 2020;20(9):981–989. doi: 10.1080/14712598.2020.1769063. [DOI] [PubMed] [Google Scholar]
  • 232.Reddy S.M., Carroll E., Nanda R. Atezolizumab for the treatment of breast cancer. Expert Rev Anticancer Ther. 2020;20(3):151–158. doi: 10.1080/14737140.2020.1732211. [DOI] [PubMed] [Google Scholar]
  • 233.Cyprian F.S., Akhtar S., Gatalica Z., Vranic S. Targeted immunotherapy with a checkpoint inhibitor in combination with chemotherapy: a new clinical paradigm in the treatment of triple-negative breast cancer. Bosn J Basic Med Sci. 2019;19(3):227–233. doi: 10.17305/bjbms.2019.4204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Ledford H. Engineered cell therapy for cancer gets thumbs up from FDA advisers. Nature. 2017;547(7663):270. doi: 10.1038/nature.2017.22304. [DOI] [PubMed] [Google Scholar]
  • 235.Schirrmacher V. From chemotherapy to biological therapy: a review of novel concepts to reduce the side effects of systemic cancer treatment (Review) Int J Oncol. 2019;54(2):407–419. doi: 10.3892/ijo.2018.4661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Vanneman M., Dranoff G. Combining immunotherapy and targeted therapies in cancer treatment. Nat Rev Cancer. 2012;12(4):237–251. doi: 10.1038/nrc3237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Hughes P.E., Caenepeel S., Wu L.C. Targeted therapy and checkpoint immunotherapy combinations for the treatment of cancer. Trends Immunol. 2016;37(7):462–476. doi: 10.1016/j.it.2016.04.010. [DOI] [PubMed] [Google Scholar]
  • 238.Grosser R., Cherkassky L., Chintala N., Adusumilli P.S. Combination immunotherapy with CAR T cells and checkpoint blockade for the treatment of solid tumors. Cancer Cell. 2019;36(5):471–482. doi: 10.1016/j.ccell.2019.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Chiossone L., Dumas P.Y., Vienne M., Vivier E. Natural killer cells and other innate lymphoid cells in cancer. Nat Rev Immunol. 2018;18(11):671–688. doi: 10.1038/s41577-018-0061-z. [DOI] [PubMed] [Google Scholar]
  • 240.Anyaegbu C.C., Lake R.A., Heel K., Robinson B.W., Fisher S.A. Chemotherapy enhances cross-presentation of nuclear tumor antigens. PLoS One. 2014;9(9):e107894. doi: 10.1371/journal.pone.0107894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Konstantinopoulos P.A., Cheng S.C., Wahner Hendrickson A.E., et al. Berzosertib plus gemcitabine versus gemcitabine alone in platinum-resistant high-grade serous ovarian cancer: a multicentre, open-label, randomised, phase 2 trial. Lancet Oncol. 2020;21(7):957–968. doi: 10.1016/S1470-2045(20)30180-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Reck M. Pembrolizumab as first-line therapy for metastatic non-small-cell lung cancer. Immunotherapy. 2018;10(2):93–105. doi: 10.2217/imt-2017-0121. [DOI] [PubMed] [Google Scholar]
  • 243.Kang C., Syed Y.Y. Atezolizumab (in combination with nab-paclitaxel):a review in advanced triple-negative breast cancer. Drugs. 2020;80(6):601–607. doi: 10.1007/s40265-020-01295-y. [DOI] [PubMed] [Google Scholar]
  • 244.Allard B., Allard D., Buisseret L., Stagg J. The adenosine pathway in immuno-oncology. Nat Rev Clin Oncol. 2020;17(10):611–629. doi: 10.1038/s41571-020-0382-2. [DOI] [PubMed] [Google Scholar]
  • 245.Pasquini S., Contri C., Borea P.A., Vincenzi F., Varani K. Adenosine and inflammation: here, there and everywhere. Int J Mol Sci. 2021;22(14):7685. doi: 10.3390/ijms22147685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Franco R., Rivas-Santisteban R., Navarro G., Reyes-Resina I. Adenosine receptor antagonists to combat cancer and to boost anti-cancer chemotherapy and immunotherapy. Cells. 2021;10(11):2831. doi: 10.3390/cells10112831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Helms R.S., Powell J.D. Rethinking the adenosine-A2AR checkpoint: implications for enhancing anti-tumor immunotherapy. Curr Opin Pharmacol. 2020;53:77–83. doi: 10.1016/j.coph.2020.07.003. [DOI] [PubMed] [Google Scholar]
  • 248.Yu F., Zhu C., Xie Q., Wang Y. Adenosine A2A receptor antagonists for cancer immunotherapy. J Med Chem. 2020;63(21):12196–12212. doi: 10.1021/acs.jmedchem.0c00237. [DOI] [PubMed] [Google Scholar]
  • 249.Yan W., Ling L., Wu Y., et al. Structure-based design of dual-acting compounds targeting adenosine A2A receptor and histone deacetylase as novel tumor immunotherapeutic agents. J Med Chem. 2021;64(22):16573–16597. doi: 10.1021/acs.jmedchem.1c01155. [DOI] [PubMed] [Google Scholar]
  • 250.Churov A., Zhulai G. Targeting adenosine and regulatory T cells in cancer immunotherapy. Hum Immunol. 2021;82(4):270–278. doi: 10.1016/j.humimm.2020.12.005. [DOI] [PubMed] [Google Scholar]
  • 251.Marks D.L., Olson R.L., Fernandez-Zapico M.E. Epigenetic control of the tumor microenvironment. Epigenomics. 2016;8(12):1671–1687. doi: 10.2217/epi-2016-0110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Lodewijk I., Nunes S.P., Henrique R., Jerónimo C., Dueñas M., Paramio J.M. Tackling tumor microenvironment through epigenetic tools to improve cancer immunotherapy. Clin Epigenet. 2021;13(1):63. doi: 10.1186/s13148-021-01046-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253.Pan X., Zheng L. Epigenetics in modulating immune functions of stromal and immune cells in the tumor microenvironment. Cell Mol Immunol. 2020;17(9):940–953. doi: 10.1038/s41423-020-0505-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Duan Q., Zhang H., Zheng J., Zhang L. Turning cold into hot: firing up the tumor microenvironment. Trends Cancer. 2020;6(7):605–618. doi: 10.1016/j.trecan.2020.02.022. [DOI] [PubMed] [Google Scholar]
  • 255.Lim W.A., June C.H. The principles of engineering immune cells to treat cancer. Cell. 2017;168(4):724–740. doi: 10.1016/j.cell.2017.01.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256.Murciano-Goroff Y.R., Warner A.B., Wolchok J.D. The future of cancer immunotherapy: microenvironment-targeting combinations. Cell Res. 2020;30(6):507–519. doi: 10.1038/s41422-020-0337-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Hulen T.M., Chamberlain C.A., Svane I.M., Met Ö. ACT up TIL now: the evolution of tumor-infiltrating lymphocytes in adoptive cell therapy for the treatment of solid tumors. Immunology. 2021;1(3):194–211. [Google Scholar]
  • 258.Newick K., O'Brien S., Moon E., Albelda S.M. CAR T cell therapy for solid tumors. Annu Rev Med. 2017;68:139–152. doi: 10.1146/annurev-med-062315-120245. [DOI] [PubMed] [Google Scholar]
  • 259.Singh A.K., McGuirk J.P. CAR T cells: continuation in a revolution of immunotherapy. Lancet Oncol. 2020;21(3):e168–e178. doi: 10.1016/S1470-2045(19)30823-X. [DOI] [PubMed] [Google Scholar]
  • 260.Jazirehi A.R. Molecular analysis of elements of melanoma insensitivity to TCR-engineered adoptive cell therapy. Int J Mol Sci. 2021;22(21):11726. doi: 10.3390/ijms222111726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261.Tan E., Gakhar N., Kirtane K. TCR gene-engineered cell therapy for solid tumors. Best Pract Res Clin Haematol. 2021;34(3):101285. doi: 10.1016/j.beha.2021.101285. [DOI] [PubMed] [Google Scholar]
  • 262.Ö Met, Jensen K.M., Chamberlain C.A., Donia M., Svane I.M. Principles of adoptive T cell therapy in cancer. Semin Immunopathol. 2019;41(1):49–58. doi: 10.1007/s00281-018-0703-z. [DOI] [PubMed] [Google Scholar]
  • 263.Wolf B., Zimmermann S., Arber C., Irving M., Trueb L., Coukos G. Safety and tolerability of adoptive cell therapy in cancer. Drug Saf. 2019;42(2):315–334. doi: 10.1007/s40264-018-0779-3. [DOI] [PubMed] [Google Scholar]
  • 264.Kaufman H.L., Maciorowski D. Advancing oncolytic virus therapy by understanding the biology. Nat Rev Clin Oncol. 2021;18(4):197–198. doi: 10.1038/s41571-021-00490-4. [DOI] [PubMed] [Google Scholar]
  • 265.Ferrucci P.F., Pala L., Conforti F., Cocorocchio E. Talimogene laherparepvec (T-VEC):an intralesional cancer immunotherapy for advanced melanoma. Cancers. 2021;13(6):1383. doi: 10.3390/cancers13061383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266.Lu L., Jiang J., Zhan M., et al. Targeting tumor-associated antigens in hepatocellular carcinoma for immunotherapy: past pitfalls and future strategies. Hepatology. 2021;73(2):821–832. doi: 10.1002/hep.31502. [DOI] [PubMed] [Google Scholar]
  • 267.Liu Z., Han C., Fu Y.X. Targeting innate sensing in the tumor microenvironment to improve immunotherapy. Cell Mol Immunol. 2020;17(1):13–26. doi: 10.1038/s41423-019-0341-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268.Bejarano L., Jordāo M.J.C., Joyce J.A. Therapeutic targeting of the tumor microenvironment. Cancer Discov. 2021;11(4):933–959. doi: 10.1158/2159-8290.CD-20-1808. [DOI] [PubMed] [Google Scholar]
  • 269.Anand U., Tudu C.K., Nandy S., et al. Ethnodermatological use of medicinal plants in India: from ayurvedic formulations to clinical perspectives - a review. J Ethnopharmacol. 2022;284:114744. doi: 10.1016/j.jep.2021.114744. [DOI] [PubMed] [Google Scholar]
  • 270.Paul S., Chakraborty S., Anand U., et al. Withania somnifera (L.) Dunal (Ashwagandha):a comprehensive review on ethnopharmacology, pharmacotherapeutics, biomedicinal and toxicological aspects. Biomed Pharmacother. 2021;143:112175. doi: 10.1016/j.biopha.2021.112175. [DOI] [PubMed] [Google Scholar]
  • 271.Lin S.R., Chang C.H., Hsu C.F., et al. Natural compounds as potential adjuvants to cancer therapy: preclinical evidence. Br J Pharmacol. 2020;177(6):1409–1423. doi: 10.1111/bph.14816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Nobili S., Lippi D., Witort E., et al. Natural compounds for cancer treatment and prevention. Pharmacol Res. 2009;59(6):365–378. doi: 10.1016/j.phrs.2009.01.017. [DOI] [PubMed] [Google Scholar]
  • 273.Mitra S., Anand U., Jha N.K., et al. Anticancer applications and pharmacological properties of piperidine and piperine: a comprehensive review on molecular mechanisms and therapeutic perspectives. Front Pharmacol. 2021;12:772418. doi: 10.3389/fphar.2021.772418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274.Mitra S., Anand U., Sanyal R., et al. Neoechinulins: molecular, cellular, and functional attributes as promising therapeutics against cancer and other human diseases. Biomed Pharmacother. 2022;145:112378. doi: 10.1016/j.biopha.2021.112378. [DOI] [PubMed] [Google Scholar]
  • 275.Bandopadhyay S., Anand U., Gadekar V.S., et al. Dioscin: a review on pharmacological properties and therapeutic values. Biofactors. 2022;48(1):22–55. doi: 10.1002/biof.1815. [DOI] [PubMed] [Google Scholar]
  • 276.Datta S., Ramamurthy P.C., Anand U., et al. Wonder or evil? : multifaceted health hazards and health benefits of Cannabis sativa and its phytochemicals. Saudi J Biol Sci. 2021;28(12):7290–7313. doi: 10.1016/j.sjbs.2021.08.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 277.Dutta T., Anand U., Saha S.C., et al. Advancing urban ethnopharmacology: a modern concept of sustainability, conservation and cross-cultural adaptations of medicinal plant lore in the urban environment. Conserv Physiol. 2021;9(1):coab073. doi: 10.1093/conphys/coab073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278.Tournilhac O., Cazin B., Leprètre S., et al. Impact of frontline fludarabine and cyclophosphamide combined treatment on peripheral blood stem cell mobilization in B-cell chronic lymphocytic leukemia. Blood. 2004;103(1):363–365. doi: 10.1182/blood-2003-05-1449. [DOI] [PubMed] [Google Scholar]
  • 279.Chu E., DeVita V.T. 15th ed. Jones & Bartlett Learning; Burlington, MA: 2015. Physician's Cancer Chemotherapy Drug Manual 2015. [Google Scholar]
  • 280.Yerram P., Reiss S.N., Modelevsky L., Gavrilovic I.T., Kaley T. Evaluation of toxicity of carmustine with or without bevacizumab in patients with recurrent or progressive high grade gliomas. J Neuro Oncol. 2019;145(1):57–63. doi: 10.1007/s11060-019-03266-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281.Karahoca M., Momparler R.L. Pharmacokinetic and pharmacodynamic analysis of 5-aza-2'-deoxycytidine (decitabine) in the design of its dose-schedule for cancer therapy. Clin Epigenet. 2013;5(1):3. doi: 10.1186/1868-7083-5-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282.Vanneste M., Huang Q., Li M., et al. High content screening identifies monensin as an EMT-selective cytotoxic compound. Sci Rep. 2019;9(1):1200. doi: 10.1038/s41598-018-38019-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 283.Rycenga H.B., Long D.T. The evolving role of DNA inter-strand crosslinks in chemotherapy. Curr Opin Pharmacol. 2018;41:20–26. doi: 10.1016/j.coph.2018.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 284.Samuels B.L., Bitran J.D. High-dose intravenous melphalan: a review. J Clin Oncol. 1995;13(7):1786–1799. doi: 10.1200/JCO.1995.13.7.1786. [DOI] [PubMed] [Google Scholar]
  • 285.Ma M.T., He M., Wang Y., et al. miR-487a resensitizes mitoxantrone (MX)-resistant breast cancer cells (MCF-7/MX) to MX by targeting breast cancer resistance protein (BCRP/ABCG2) Cancer Lett. 2013;339(1):107–115. doi: 10.1016/j.canlet.2013.07.016. [DOI] [PubMed] [Google Scholar]
  • 286.Graham J., Muhsin M., Kirkpatrick P. Oxaliplatin. Nat Rev Drug Discov. 2004;3(1):11–12. doi: 10.1038/nrd1287. [DOI] [PubMed] [Google Scholar]
  • 287.Jacinto F.V., Esteller M. MGMT hypermethylation: a prognostic foe, a predictive friend. DNA Repair. 2007;6(8):1155–1160. doi: 10.1016/j.dnarep.2007.03.013. [DOI] [PubMed] [Google Scholar]
  • 288.Arrieta O., Lara-Mejía L., Zatarain-Barrón Z.L. Carboplatin plus etoposide or topotecan for small-cell lung cancer. Lancet Oncol. 2020;21(9):1132–1134. doi: 10.1016/S1470-2045(20)30427-7. [DOI] [PubMed] [Google Scholar]
  • 289.Hwang D.Y., Kim S.J., Cheong J.W., et al. High pre-transplant serum ferritin and busulfan-thiotepa conditioning regimen as risk factors for hepatic sinusoidal obstructive syndrome after autologous stem cell transplantation in patients with malignant lymphoma. Leuk Lymphoma. 2016;57(1):51–57. doi: 10.3109/10428194.2015.1041387. [DOI] [PubMed] [Google Scholar]
  • 290.D'Incalci M., Badri N., Galmarini C.M., Allavena P. Trabectedin, a drug acting on both cancer cells and the tumour microenvironment. Br J Cancer. 2014;111(4):646–650. doi: 10.1038/bjc.2014.149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291.Maltzman J.S., Koretzky G.A. Azathioprine: old drug, new actions. J Clin Invest. 2003;111(8):1122–1124. doi: 10.1172/JCI18384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292.Hasanali Z.S., Saroya B.S., Stuart A., et al. Epigenetic therapy overcomes treatment resistance in T cell prolymphocytic leukemia. Sci Transl Med. 2015;7(293):293ra102. doi: 10.1126/scitranslmed.aaa5079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 293.Bonate P.L., Arthaud L., Cantrell W.R., Stephenson K., Secrist J.A., Weitman S. Discovery and development of clofarabine: a nucleoside analogue for treating cancer. Nat Rev Drug Discov. 2006;5(10):855–863. doi: 10.1038/nrd2055. [DOI] [PubMed] [Google Scholar]
  • 294.Ricci F., Tedeschi A., Morra E., Montillo M. Fludarabine in the treatment of chronic lymphocytic leukemia: a review. Therapeut Clin Risk Manag. 2009;5(1):187–207. doi: 10.2147/tcrm.s3688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295.Mini E., Nobili S., Caciagli B., Landini I., Mazzei T. Cellular pharmacology of gemcitabine. Ann Oncol. 2006;17(Suppl 5):v7–v12. doi: 10.1093/annonc/mdj941. [DOI] [PubMed] [Google Scholar]
  • 296.Engels F.K., Sparreboom A., Mathot R.A.A., Verweij J. Potential for improvement of docetaxel-based chemotherapy: a pharmacological review. Br J Cancer. 2005;93(2):173–177. doi: 10.1038/sj.bjc.6602698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 297.Cohen M.H., Johnson J.R., Massie T., et al. Approval summary: nelarabine for the treatment of T-cell lymphoblastic leukemia/lymphoma. Clin Cancer Res. 2006;12(18):5329–5335. doi: 10.1158/1078-0432.CCR-06-0606. [DOI] [PubMed] [Google Scholar]
  • 298.Cannon T., Mobarek D., Wegge J., Tabbara I.A. Hairy cell leukemia: current concepts. Cancer Invest. 2008;26(8):860–865. doi: 10.1080/07357900801965034. [DOI] [PubMed] [Google Scholar]
  • 299.Wang H., Wang Y. LC-MS/MS coupled with stable isotope dilution method for the quantification of 6-thioguanine and S6-methylthioguanine in genomic DNA of human cancer cells treated with 6-thioguanine. Anal Chem. 2010;82(13):5797–5803. doi: 10.1021/ac1008628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 300.Sertel S., Tome M., Briehl M.M., et al. Factors determining sensitivity and resistance of tumor cells to arsenic trioxide. PLoS One. 2012;7(5):e35584. doi: 10.1371/journal.pone.0035584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 301.Madaan K., Kaushik D., Verma T. Hydroxyurea: a key player in cancer chemotherapy. Expert Rev Anticancer Ther. 2012;12(1):19–29. doi: 10.1586/era.11.175. [DOI] [PubMed] [Google Scholar]
  • 302.Scripture C.D., Szebeni J., Loos W.J., Figg W.D., Sparreboom A. Comparative in vitro properties and clinical pharmacokinetics of Paclitaxel following the administration of taxol(r) and paxene(r) Cancer Biol Ther. 2005;4(5):555–560. doi: 10.4161/cbt.4.5.1664. [DOI] [PubMed] [Google Scholar]
  • 303.Alken S., Kelly C.M. Benefit risk assessment and update on the use of docetaxel in the management of breast cancer. Cancer Manag Res. 2013;5:357–365. doi: 10.2147/CMAR.S49321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 304.Chan E.S., Cronstein B.N. Mechanisms of action of methotrexate. Bull Hosp Jt Dis. 2013;71(Suppl 1):S5–S8. [PubMed] [Google Scholar]
  • 305.Adjei A.A. Pharmacology and mechanism of action of pemetrexed. Clin Lung Cancer. 2004;5(Suppl 2):S51–S55. doi: 10.3816/clc.2004.s.003. [DOI] [PubMed] [Google Scholar]
  • 306.Weatherstone K., Meyer T. Streptozocin-based chemotherapy is not history in neuroendocrine tumours. Targeted Oncol. 2012;7(3):161–168. doi: 10.1007/s11523-012-0224-y. [DOI] [PubMed] [Google Scholar]
  • 307.Haque A., Rahman M.A., Faizi M.S.H., Khan M.S. Next generation antineoplastic agents: a review on structurally modified vinblastine (VBL) analogues. Curr Med Chem. 2018;25(14):1650–1662. doi: 10.2174/0929867324666170502123639. [DOI] [PubMed] [Google Scholar]
  • 308.Beaver C. Vincristine minibag administration: a quality improvement project to minimize medical errors. Clin J Oncol Nurs. 2018;22(6):669–672. doi: 10.1188/18.CJON.669-672. [DOI] [PubMed] [Google Scholar]
  • 309.Mehta-Shah N., Ratner L., Horwitz S.M. Adult T-cell leukemia/lymphoma. J Oncol Pract. 2017;13(8):487–492. doi: 10.1200/JOP.2017.021907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 310.Altinoz M.A., Ozpinar A., Alturfan E.E., Elmaci I. Vinorelbine's anti-tumor actions may depend on the mitotic apoptosis, autophagy and inflammation: hypotheses with implications for chemo-immunotherapy of advanced cancers and pediatric gliomas. J Chemother. 2018;30(4):203–212. doi: 10.1080/1120009X.2018.1487149. [DOI] [PubMed] [Google Scholar]
  • 311.Jäger U., Barcellini W., Broome C.M., et al. Diagnosis and treatment of autoimmune hemolytic Anemia in adults: recommendations from the first international consensus meeting. Blood Rev. 2020;41:100648. doi: 10.1016/j.blre.2019.100648. [DOI] [PubMed] [Google Scholar]
  • 312.Kreitman R.J., Arons E. Update on hairy cell leukemia. Clin Adv Hematol Oncol. 2018;16(3):205–215. [PMC free article] [PubMed] [Google Scholar]
  • 313.Begna K., Abdelatif A., Schwager S., Hanson C., Pardanani A., Tefferi A. Busulfan for the treatment of myeloproliferative neoplasms: the Mayo Clinic experience. Blood Cancer J. 2016;6(5):e427. doi: 10.1038/bcj.2016.34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 314.Palmer J., McCune J.S., Perales M.A., et al. Personalizing busulfan-based conditioning: considerations from the American society for blood and marrow transplantation practice guidelines committee. Biol Blood Marrow Transplant. 2016;22(11):1915–1925. doi: 10.1016/j.bbmt.2016.07.013. [DOI] [PubMed] [Google Scholar]
  • 315.Ho G.Y., Woodward N., Coward J.I.G. Cisplatin versus carboplatin: comparative review of therapeutic management in solid malignancies. Crit Rev Oncol Hematol. 2016;102:37–46. doi: 10.1016/j.critrevonc.2016.03.014. [DOI] [PubMed] [Google Scholar]
  • 316.Lee H.Z., Miller B.W., Kwitkowski V.E., et al. U.S. food and drug administration approval: obinutuzumab in combination with chlorambucil for the treatment of previously untreated chronic lymphocytic leukemia. Clin Cancer Res. 2014;20(15):3902–3907. doi: 10.1158/1078-0432.CCR-14-0516. [DOI] [PubMed] [Google Scholar]
  • 317.Hew J., Pham D., Matthews Hew T., Minocha V. A novel treatment with obinutuzumab-chlorambucil in a patient with B-cell prolymphocytic leukemia: a case report and review of the literature. J Investig Med High Impact Case Rep. 2018;6 doi: 10.1177/2324709618788674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 318.Makovec T. Cisplatin and beyond: molecular mechanisms of action and drug resistance development in cancer chemotherapy. Radiol Oncol. 2019;53(2):148–158. doi: 10.2478/raon-2019-0018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 319.Raudenska M., Balvan J., Fojtu M., Gumulec J., Masarik M. Unexpected therapeutic effects of cisplatin. Metallomics. 2019;11(7):1182–1199. doi: 10.1039/c9mt00049f. [DOI] [PubMed] [Google Scholar]
  • 320.Casak S.J., Lemery S.J., Shen Y.L., et al. U.S. Food and drug administration approval: rituximab in combination with fludarabine and cyclophosphamide for the treatment of patients with chronic lymphocytic leukemia. Oncol. 2011;16(1):97–104. doi: 10.1634/theoncologist.2010-0306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 321.Jiang G., Li R.H., Sun C., Liu Y.Q., Zheng J.N. Dacarbazine combined targeted therapy versus dacarbazine alone in patients with malignant melanoma: a meta-analysis. PLoS One. 2014;9(12):e111920. doi: 10.1371/journal.pone.0111920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 322.Velho T.R. Metastatic melanoma - a review of current and future drugs. Drugs Context. 2012;2012:212242. doi: 10.7573/dic.212242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 323.Krauss A.C., Gao X., Li L., et al. FDA approval summary: (daunorubicin and cytarabine) liposome for injection for the treatment of adults with high-risk acute myeloid leukemia. Clin Cancer Res. 2019;25(9):2685–2690. doi: 10.1158/1078-0432.CCR-18-2990. [DOI] [PubMed] [Google Scholar]
  • 324.Blair H.A. Daunorubicin/cytarabine liposome: a review in acute myeloid leukaemia. Drugs. 2018;78(18):1903–1910. doi: 10.1007/s40265-018-1022-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 325.Dhillon S. Decitabine/cedazuridine: first approval. Drugs. 2020;80(13):1373–1378. doi: 10.1007/s40265-020-01389-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 326.Kanwal U., Irfan Bukhari N., Ovais M., Abass N., Hussain K., Raza A. Advances in nano-delivery systems for doxorubicin: an updated insight. J Drug Target. 2018;26(4):296–310. doi: 10.1080/1061186X.2017.1380655. [DOI] [PubMed] [Google Scholar]
  • 327.Cagel M., Grotz E., Bernabeu E., Moretton M.A., Chiappetta D.A. Doxorubicin: nanotechnological overviews from bench to bedside. Drug Discov Today. 2017;22(2):270–281. doi: 10.1016/j.drudis.2016.11.005. [DOI] [PubMed] [Google Scholar]
  • 328.Buzdar A.U., Suman V.J., Meric-Bernstam F., et al. Fluorouracil, epirubicin, and cyclophosphamide (FEC-75) followed by paclitaxel plus trastuzumab versus paclitaxel plus trastuzumab followed by FEC-75 plus trastuzumab as neoadjuvant treatment for patients with HER2-positive breast cancer (Z1041):a randomised, controlled, phase 3 trial. Lancet Oncol. 2013;14(13):1317–1325. doi: 10.1016/S1470-2045(13)70502-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 329.Hollingshead L.M., Faulds D. Idarubicin. A review of its pharmacodynamic and pharmacokinetic properties, and therapeutic potential in the chemotherapy of cancer. Drugs. 1991;42(4):690–719. doi: 10.2165/00003495-199142040-00010. [DOI] [PubMed] [Google Scholar]
  • 330.Matz E.L., Hsieh M.H. Review of advances in uroprotective agents for cyclophosphamide- and ifosfamide-induced hemorrhagic cystitis. Urology. 2017;100:16–19. doi: 10.1016/j.urology.2016.07.030. [DOI] [PubMed] [Google Scholar]
  • 331.de Man F.M., Goey A.K.L., van Schaik R.H.N., Mathijssen R.H.J., Bins S. Individualization of irinotecan treatment: a review of pharmacokinetics, pharmacodynamics, and pharmacogenetics. Clin Pharmacokinet. 2018;57(10):1229–1254. doi: 10.1007/s40262-018-0644-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 332.KuKanich B., Warner M., Hahn K. Analysis of lomustine drug content in FDA-approved and compounded lomustine capsules. J Am Vet Med Assoc. 2017;250(3):322–326. doi: 10.2460/javma.250.3.322. [DOI] [PubMed] [Google Scholar]
  • 333.Sun J., Wei Q., Zhou Y., Wang J., Liu Q., Xu H. A systematic analysis of FDA-approved anticancer drugs. BMC Syst Biol. 2017;11(Suppl 5):87. doi: 10.1186/s12918-017-0464-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 334.Fox E.J. Mechanism of action of mitoxantrone. Neurology. 2004;63(12 Suppl 6):S15–S18. doi: 10.1212/wnl.63.12_suppl_6.s15. [DOI] [PubMed] [Google Scholar]
  • 335.Ibrahim A., Hirschfeld S., Cohen M.H., Griebel D.J., Williams G.A., Pazdur R. FDA drug approval summaries: oxaliplatin. Oncol. 2004;9(1):8–12. doi: 10.1634/theoncologist.9-1-8. [DOI] [PubMed] [Google Scholar]
  • 336.Stein A., Arnold D. Oxaliplatin: a review of approved uses. Expet Opin Pharmacother. 2012;13(1):125–137. doi: 10.1517/14656566.2012.643870. [DOI] [PubMed] [Google Scholar]
  • 337.Mutter N., Stupp R. Temozolomide: a milestone in neuro-oncology and beyond? Expert Rev Anticancer Ther. 2006;6(8):1187–1204. doi: 10.1586/14737140.6.8.1187. [DOI] [PubMed] [Google Scholar]
  • 338.Ardizzoni A., Hansen H., Dombernowsky P., et al. Topotecan, a new active drug in the second-line treatment of small-cell lung cancer: a phase II study in patients with refractory and sensitive disease. The European organization for research and treatment of cancer early clinical studies group and new drug development office, and the lung cancer cooperative group. J Clin Oncol. 1997;15(5):2090–2096. doi: 10.1200/JCO.1997.15.5.2090. [DOI] [PubMed] [Google Scholar]
  • 339.Garst J. Topotecan: an evolving option in the treatment of relapsed small cell lung cancer. Therapeut Clin Risk Manag. 2007;3(6):1087–1095. [PMC free article] [PubMed] [Google Scholar]
  • 340.Eckardt J.R., von Pawel J., Pujol J.L., et al. Phase III study of oral compared with intravenous topotecan as second-line therapy in small-cell lung cancer. J Clin Oncol. 2007;25(15):2086–2092. doi: 10.1200/JCO.2006.08.3998. [DOI] [PubMed] [Google Scholar]
  • 341.Kokolo M.B., Fergusson D., O'Neill J., et al. Effectiveness and safety of thiotepa as conditioning treatment prior to stem cell transplant in patients with central nervous system lymphoma. Leuk Lymphoma. 2014;55(12):2712–2720. doi: 10.3109/10428194.2014.889825. [DOI] [PubMed] [Google Scholar]
  • 342.Barone A., Chi D.C., Theoret M.R., et al. FDA approval summary: trabectedin for unresectable or metastatic liposarcoma or leiomyosarcoma following an anthracycline-containing regimen. Clin Cancer Res. 2017;23(24):7448–7453. doi: 10.1158/1078-0432.CCR-17-0898. [DOI] [PubMed] [Google Scholar]
  • 343.Rosenberg J.D., Burian C., Waalen J., Saven A. Clinical characteristics and long-term outcome of young hairy cell leukemia patients treated with cladribine: a single-institution series. Blood. 2014;123(2):177–183. doi: 10.1182/blood-2013-06-508754. [DOI] [PubMed] [Google Scholar]
  • 344.Zhenchuk A., Lotfi K., Juliusson G., Albertioni F. Mechanisms of anti-cancer action and pharmacology of clofarabine. Biochem Pharmacol. 2009;78(11):1351–1359. doi: 10.1016/j.bcp.2009.06.094. [DOI] [PubMed] [Google Scholar]
  • 345.Schmiegelow K., Al-Modhwahi I., Andersen M.K., et al. Methotrexate/6-mercaptopurine maintenance therapy influences the risk of a second malignant neoplasm after childhood acute lymphoblastic leukemia: results from the NOPHO ALL-92 study. Blood. 2009;113(24):6077–6084. doi: 10.1182/blood-2008-11-187880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 346.Dillman R.O. Pentostatin (Nipent®) in the treatment of chronic lymphocyte leukemia and hairy cell leukemia. Expert Rev Anticancer Ther. 2004;4(1):27–36. doi: 10.1586/14737140.4.1.27. [DOI] [PubMed] [Google Scholar]
  • 347.Munshi P.N., Lubin M., Bertino J.R. 6-thioguanine: a drug with unrealized potential for cancer therapy. Oncol. 2014;19(7):760–765. doi: 10.1634/theoncologist.2014-0178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 348.Dent S., Messersmith H., Trudeau M. Gemcitabine in the management of metastatic breast cancer: a systematic review. Breast Cancer Res Treat. 2008;108(3):319–331. doi: 10.1007/s10549-007-9610-z. [DOI] [PubMed] [Google Scholar]
  • 349.Moysan E., Bastiat G., Benoit J.P. Gemcitabine versus Modified Gemcitabine: a review of several promising chemical modifications. Mol Pharm. 2013;10(2):430–444. doi: 10.1021/mp300370t. [DOI] [PubMed] [Google Scholar]
  • 350.Matt P., van Zwieten-Boot B., Calvo Rojas G., et al. The European Medicines Agency review of Tegafur/Gimeracil/Oteracil (Teysuno™) for the treatment of advanced gastric cancer when given in combination with cisplatin: summary of the Scientific Assessment of the Committee for medicinal products for human use (CHMP) Oncol. 2011;16(10):1451–1457. doi: 10.1634/theoncologist.2011-0224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 351.Alimoghaddam K. A review of arsenic trioxide and acute promyelocytic leukemia. Int J Hematol Oncol Stem Cell Res. 2014;8(3):44–54. [PMC free article] [PubMed] [Google Scholar]
  • 352.Emadi A., Gore S.D. Arsenic trioxide - an old drug rediscovered. Blood Rev. 2010;24(4–5):191–199. doi: 10.1016/j.blre.2010.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 353.Menzin A.W., King S.A., Aikins J.K., Mikuta J.J., Rubin S.C. Taxol (paclitaxel) was approved by FDA for the treatment of patients with recurrent ovarian cancer. Gynecol Oncol. 1994;54(1):103. [PubMed] [Google Scholar]
  • 354.Li J., Ren J., Sun W. Systematic review of ixabepilone for treating metastatic breast cancer. Breast Cancer. 2017;24(2):171–179. doi: 10.1007/s12282-016-0717-0. [DOI] [PubMed] [Google Scholar]
  • 355.Abolmaali S.S., Tamaddon A.M., Dinarvand R. A review of therapeutic challenges and achievements of methotrexate delivery systems for treatment of cancer and rheumatoid arthritis. Cancer Chemother Pharmacol. 2013;71(5):1115–1130. doi: 10.1007/s00280-012-2062-0. [DOI] [PubMed] [Google Scholar]
  • 356.Moore A.S., Nelson R.W., Henry C.J., et al. Streptozocin for treatment of pancreatic islet cell tumors in dogs: 17 cases (1989-1999) J Am Vet Med Assoc. 2002;221(6):811–818. doi: 10.2460/javma.2002.221.811. [DOI] [PubMed] [Google Scholar]
  • 357.Said R., Tsimberidou A.M. Pharmacokinetic evaluation of vincristine for the treatment of lymphoid malignancies. Expet Opin Drug Metabol Toxicol. 2014;10(3):483–494. doi: 10.1517/17425255.2014.885016. [DOI] [PubMed] [Google Scholar]
  • 358.Martino E., Casamassima G., Castiglione S., et al. Vinca alkaloids and analogues as anti-cancer agents: looking back, peering ahead. Bioorg Med Chem Lett. 2018;28(17):2816–2826. doi: 10.1016/j.bmcl.2018.06.044. [DOI] [PubMed] [Google Scholar]
  • 359.Smith G.A. Current status of vinorelbine for breast cancer. Oncology (Williston Park) 1995;9(8):767–773. discussion 774, 776, 779. [PubMed] [Google Scholar]
  • 360.Gasparini G., Caffo O., Barni S., et al. Vinorelbine is an active antiproliferative agent in pretreated advanced breast cancer patients: a phase II study. J Clin Oncol. 1994;12(10):2094–2101. doi: 10.1200/JCO.1994.12.10.2094. [DOI] [PubMed] [Google Scholar]

Articles from Genes & Diseases are provided here courtesy of Chongqing Medical University

RESOURCES