3.1. Intrinsic Drug Resistance
The natural resistance that occurs before the patient is exposed to medications is typically referred to as intrinsic resistance and can influence treatment effectiveness. The latter can be brought on by several factors, such as increased DNA repair capacity, altered drug metabolism, mutated or altered drug targets, reduced drug accumulation and deactivated cell death signals [
11,
12].
Cancer stem cells (CSCs) exhibit drug resistance because they overexpress adenosine triphosphate (ATP)-binding cassette (ABC) transporters [
16]. Through certain regulatory genes, FOXM1, a transcription factor specifically for cell proliferation, controls the transition between the G1/S and G2/M cell cycle phases. Additionally, it is an oncogene that promotes the expansion and multiplication of cancer cells [
17]. Through the expression of ABCC5 (ATP binding cassette subfamily C member 5), FOXM1 overexpression causes paclitaxel resistance in nasopharyngeal carcinoma [
18]. Growth differentiation factor-15 (GDF-15) is a member of the superfamily of transforming growth factor-beta (TGF-β). Proliferation, angiogenesis, stemness, metastasis, drug resistance, and immunological modulation are all associated with overexpression of GDF-15 in cancer. It was demonstrated that stemness and indicators of treatment resistance were significantly positively correlated with GDF-15 expression in breast cancer patients. This suggests that the p-Akt/FOXM1 axis mediates the relationship between increased GDF-15 expression and enhanced stemness and treatment resistance in breast cancer [
19]. Oestrogen receptor positive (ER+)/ human epidermal growth factor receptor 2 positive (HER2+) breast cancer is strongly influenced by the HER2-E subtype and erbB2, which results in resistance to endocrine therapy and a higher probability of recurrence [
20]. About 20–30% of metastatic breast tumours overexpress the human epidermal growth factor receptor 2 (HER2/erbB2), which is associated with a poor prognosis [
21]. In studies using trastuzumab as the sole treatment, over two thirds of patients showed intrinsic resistance to the drug [
22,
23]. High levels of GDF15 may be a factor in trastuzumab resistance in HER2 overexpressing breast cancer cells through activation of TGF-β receptor-Src-HER2 signalling crosstalk [
24]. Furthermore, the aberrant activation of the phosphoinositol-3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/Akt/mTOR) signalling pathway is closely related to resistance to anti-HER2 treatment [
25]. Moreover, due to the genetic mutation(s) of genes involved in cancer cell proliferation and/or death, intrinsic drug resistance may develop in cancer cells before therapy. For instance, HER2 overexpression induced EMT and promoted resistance to cisplatin in gastric cancer cells [
26]. CSCs and EMT are both associated to intrinsic drug resistance via these concurrent alterations mentioned above [
27,
28].
Intercellular genetic heterogeneity in cancer can result from genomic instability, which is characterised by mutations, gene amplifications, chromosomal rearrangements, gene deletions, gene translocations and alterations in microRNA [
29]. Moreover, genotypic changes can have an impact on epigenetic variables affecting the heterogeneity of the mRNA, transcriptome, and proteome [
30].
3.2. Acquired Drug Resistance
Gradual declines in a drug's ability to treat cancer after treatment can indicate acquired resistance. A number of factors can contribute to acquired resistance, including changes in the TME following therapy through various mechanisms, such as low pH, hypoxia, shifts and polarisations in the immune cell population, exosomes, various secretomes, vascular abnormalities, and soluble factors derived from stromal cells [
11,
12,
31]. Paracrine signalling connections between stromal and tumour cells, mutations or altered levels of drug target expression and activation of a second proto-oncogene that develops into the driver gene, can also contribute to acquired dug resistance [
11,
12,
32].
Targeted medicines cause subtler alterations that can be classified as acquired resistance after repeated exposures or early adaptive responses. Adaptive responses may be the cause of transient clinical reactions because they might happen so quickly that no response is ever clinically evident. Adaptive processes are frequently the result of epigenetic modification and/or non-genetic relief of negative feedback of signalling pathways, which activates parallel pathways or reactivates the initial one [
33,
34]. For example, due to the reactivation of upstream receptor tyrosine kinases (RTKs), such as epidermal growth factor receptor (EGFR), BRAF-mutant colorectal tumours are resistant to BRAF inhibitors, while low level of EGFR expression in BRAF-mutant melanomas were not affected by the negative feedback relief [
35,
36].
New genetic mutations can cause resistance and regeneration in cancers that had previously shrunk. Whole-genome sequencing comparing the genetic profiles of eight patients with acute myeloid leukaemia before and after relapse revealed novel gene mutations (e.g., DAXX, DDX41, DIS3, SMC3 and WAC) responsible for tumour resistance and regeneration [
37]. Chemotherapeutic medications disrupt malignant cells' DNA, which probably accelerates the occurrence of new mutations. Also linked to acquired chemoresistance is the crosstalk that occurs between tumour cells and their microenvironment as the disease progresses [
38]. This will be discussed later in the TME section below (section 3.10).
Some non-small-cell lung cancer (NSCLC) patients experience acquired resistance due to circumstances that can interfere with EGFR signalling, such as the upregulation of other RTKs like MET, the downstream activation of specific pathway elements, or phenotypic and histological changes [
39]. Recently it was demonstrated that EGFR signalling pathways was activated by autocrine EGF and TGF-α and withstand c-Met and anaplastic lymphoma kinase (ALK) inhibition leading to primary and acquired resistance to TAE684/SGX-523 (ALK/c-Met inhibitors) in NSCLC [
40]. In hepatocellular carcinoma (HCC) cells that heavily express c-MET, hepatocyte growth factor (HGF) activated the downstream PI3K/Akt and mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) pathways through c-MET and concurrently reduced the anticancer effects of lenvatinib (a tyrosine kinase inhibitor) and promoted EMT [
41]. Activating PIK3CA mutations in HER2+ breast cancer will unable a favourable response to pyrotinib plus trastuzumab neoadjuvant therapy [
42]. By encouraging FOXD1 translation through PIK3CA/PI3K/Akt/mTOR signalling, FOXD1-AS1 (an oncogenic long non-coding RNAs (lncRNA)) exacerbates gastric cancer development and chemoresistance [
43]. Moreover, one of the primary causes of medication resistance is the point mutation in the c-ros oncogene 1 (ROS1) gene. ROS1 is a receptor of the insulin family of tyrosine kinases. Recently it was demonstrated that the point mutations CD74-ROS1 D2033N and CD74-ROS1 S1986F render NSCLC cells resistant to crizotinib via the FAK/PI3K/Akt signalling pathway activation [
44].
Activation of hypoxia-inducible pathways, EMT, interaction between the PI3K/Akt and Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathways, and enrichment of tumour-initiating cell population are some of the processes that cause acquired resistance to sorafenib [
45]. Furthermore, potential mechanisms were revealed that underlie acquired resistance to gemcitabine in gallbladder cancer such as disruption of drug metabolism, and activation of receptor and non RTK (
i.e., PDGFRA, ABL1 and LYN) as well as the increased expression of EMT-related markers, FN1, CDA and LAMC2 [
46].
A significant increase was observed in single-nucleotide variants in the genes ATM, ATR, BRCA1, LRP1B, MAP2K1, PIK3CG and ZNF217 in addition to BRAF, KRAS, NRAS and EGFR among tumours receiving prior anti-EGFR [
47]. Genes with possible signalling implications and those involved in DNA repair pathways made up the two main categories of these changes. ZNF217 and PIK3CG converge on Akt1 signalling which may encourage anti-EGFR acquired resistance. Protein kinase MAP2K1, whose acquisition occurs following anti-EGFR therapy, enhances the translation of signalling from MEK to ERK [
48,
49]. LRP1B inhibits β-catenin signalling [
50]. β-catenin activation might be a different route to get around EGFR suppression, similar to Akt1 bypass signalling. It is interesting to note that after exposure to anti-EGFR, a greater frequency of mutations in BRCA1, ATR and ATM was found. These modifications could aid in the evolution of a response to targeted therapy and could also account for increases in relative tumour mutation burden (rTMB) in patients exposed to anti-EGFR leading to acquired resistance [
47].
Activation of EGF like domain multiple 7 (EGFL7)/Notch signalling in lung cancer cells trigger resistance to EGFR inhibitors [
51]. Furthermore, SNHG7 (a lncRNA) activate the Notch1/Jagged1/HES1 pathway resulting in tumour cell stemness and resistance to folfirinox in pancreatic cancer cells [
52].
3.3. Altered drug targets
One of the main causes of drug resistance is drug targeting alteration, which occurs when drug targets' expression and functionality are altered. Di(2-ethylhexyl) phthalate (DEHP) is a chemical that is frequently found in everyday items and polyvinylchloride medical equipment. As a result, phthalates can enter the human body through eating, inhalation, and medical procedures. Phthalates induce cancer progression and chemotherapeutic resistance [
53]. Recently it was demonstrated that DEHP increased trefoil factor 3 (TFF3) expression through the vinculin/aryl hydrocarbon receptor (AhR)/ERK signalling pathway which induced EMT [
54]. In human breast cancer, the expression of the oncogene TFF3 is favourably connected with both ER+ and negative cells, and it increases cell metastasis, invasion, proliferation and treatment resistance [
55]. Through the ubiquitination pathway, DEHP promoted AhR-related changes in oestrogen receptor expression, which reduced tamoxifen's effects in AhR knockout mice [
54].
T315I point mutation that arises in BCR-ABL kinase domain is the most frequent mutation in BCR-ABL that causes resistance to first-generation (imatinib) or second-generation tyrosine kinase inhibitors (TKIs) that target the BCR-ABL protein, leading to a poor clinical prognosis in chronic myeloid leukaemia [
56,
57]. Due to the activation of intrinsic signalling pathways such as the RAS/RAF/MAPK/ERK, GSK3 β and JAK/STAT5 pathways, imatinib intolerance or initial resistance arises, and many leukaemic patients acquire secondary resistance [
58,
59,
60,
61]. Most cellular intrinsic mechanisms play a role in the development of resistance; either directly through BCR-ABL1 point mutations, which predominate in primary resistance, or indirectly through activation of signalling pathways independent of BCR-ABL1, which frequently lead to disease recurrence and therapy relapse [
59,
62]. Typically, such activation frequently happens in a BCR-ABL1-independent manner; as a result, those oncogenic pathways continue to be active even after treating leukaemic cells with imatinib, including nonmutated BCR-ABL1 cells.
Moreover, osimertinib is a third generation powerful EGFR-TKI used to treat NSCLC patients with EGFR mutations. The therapeutic use of osimertinib is nonetheless restricted by the emergence of acquired resistance associated with the triple mutation Del19/T790M/C797S in EGFR [
63]. Furthermore, mutations at either V550 (a gatekeeper residue) or C552 (hinge-1 residue) in the kinase domain of fibroblast growth factor receptor 4 (FGFR4) prevent fisogatinib (a potent and selective FGFR4 inhibitor) from interacting with the FGFR4's ATP binding site resulting in acquired clinical resistance to fisogatinib in patients with HCC [
64].
TKIs can prevent downstream pathways from being activated improperly by aberrant protein tyrosine kinases (PTKs). PI3K/Akt, RAS/MAPK/ERK, and JAK/STAT are examples of key signalling pathways that control a variety of cellular processes by stimulating proliferation, encouraging angiogenesis, preventing apoptosis, and promoting drug resistance [
65]. Therefore, due to mutations at the drug binding sites, TKIs lose the ability to inhibit PTKs (e.g., FGFRs, EGFRs, ALK, platelet-derived growth factor receptor (PDGFRs), Insulin-like growth factor receptor (IGFRs), vascular endothelial growth factor receptor (VEGFRs)), resulting in constant activation of downstream signalling pathways.
3.7. Epigenetics modifications
Cell destiny and pathogenic provenience are greatly influenced by epigenetics. It appears that non-genetic heterogeneity contributes to the development of cancer-causing cells and/or resistance to treatment. Impairment in gene expression is caused by epigenetic alterations, which last for several cell divisions and finally result in non-genetic heterogeneity and treatment resistance [
105]. The development of chemoresistance in cancer is fuelled by epigenetic changes that are linked to histone modification, DNA methylation, chromatin remodelling, and changes associated to non-coding RNA (ncRNAs) [
106]. Accumulating evidence show that epigenetic changes contribute to the development of various resistance mechanisms, such as improved DNA repair, enhanced drug efflux, and defective apoptosis. For example, the chromodomain helicase DNA-binding protein 4 (CHD4), which modulates chromatin remodelling, specifically causes drug resistance in breast cancer gene1/2 (BRCA1/2) deficient cells through aiding DNA damage repair [
107]. Recently, it was demonstrated that by interacting with major vault protein (MVP), CHD4 encourages gastric cancer cell proliferation and chemoresistance. As well as by stimulating drug efflux, CHD4 promotes the reduction in the intracellular concentration of cisplatin. It also enhances the protein interaction between ERK1/2 and MEK1/2 leading to the activation of MVP/MEK/ERK signalling axis [
108].
Moreover, DNA methylation and gene expression profiles of fulvestrant- and tamoxifen-resistant MCF7 derivatives with oestrogen-responsive MCF7 human breast cancer cells were analysed. Resistance to tamoxifen is developed by significant alterations in downstream ER target gene networks, whereas acquired resistance to fulvestrant revealed a general up-regulation of growth-stimulatory pathways including cytokines and cytokine receptors, the EGFR, ErbB2 and related proteins, the Notch pathway, the Wnt/β-catenin pathway and the interferons (IFN) signalling pathway/IFN-inducible genes were among the prominently altered pathways in MCF7 cells resistant to fulvestrant [
109]. For instance, demethylation of DNA near an oncogene's promoter region would increase the gene's expression, leading to treatment resistance. In a resistant HCC cell line, thymosin 4 (Tβ4), a G-actin monomer binding protein, was shown to be enhanced through DNA demethylation and active modification of histone H3 at the promoter region [
110]. In the HCC cell line, overexpression of Tβ4 caused stem cell-like capabilities to develop, as well as in vivo resistance to the VEGFR inhibitor sorafenib [
110].
A study shows that sorafenib resistance develops because of the histone demethylase KDM1A also known as Lysine demethylase 1A (LSD1). They found that cells resistant to sorafenib (a TKIs) had a higher capacity for self-renewal. KDM1A's importance for the stemness of liver CSCs by epigenetic alteration was previously discovered by the same team [
111]. They found a potential mechanism by which KDM1A causes resistance to sorafenib through the control of important β-catenin signalling pathway antagonists. They also showed that KDM1A is necessary for maintaining the stemness of resistant HCC cells to sorafenib [
111].
The first histone modification enzyme shown to be linked to drug resistance to several anticancer drugs is a lysine demethylase called Lysine demethylase 5A (KDM5A) [
112,
113]. Erlotinib (an EGFR inhibitor) was less effective against breast cancer cells with amplifications of the KDM5A gene due to increased expression of a set of genes associated with apoptosis/cell cycle, including the apoptosis effector BCL2 antagonist/killer1 (BAK1) and the tumour suppressor p21 [
113]. Lewis lung carcinoma and renal cell carcinoma become resistant to sunitinib (a RTK inhibitor) due to KDM5C which was discovered to be a significant epigenetic regulator in this process. In patients with NSCLC, KDM1A is crucial for inducing gefitinib resistance by the development of hypoxia through generating EMT [
114].
Moreover, by changing gene expression and the structure of regulatory proteins, N6-methyladenosine (m6A), a particular type of RNA alteration, influences the development of tumours [
115]. KIAA1429 plays a vital role in m6A methylation or controls ncRNAs, including microRNAs (miRNAs) and lncRNA to promote the growth and metastasis of many cancers [
116,
117]. Recently it was demonstrated that the activation of the JNK/MAPK signalling pathway results in m6A KIAA1429-mediated gefitinib resistance in lung adenocarcinoma cells [
118].
The effects of mucin 17 (MUC17) on the epigenome of EGFR-TKI-acquired drug resistance was examined in NSCLC cells. Gefitinib/osimertinib-resistant (GR/OR) cells were found to increase genome-wide DNA hypermethylation, notably in 5-UTR related to several oncogenic pathways, where GR/OR cells had a pro-oncogenic effect by decreasing MUC17 expression. The downregulation of MUC17 caused by acquired GR/OR was triggered by a methylation promoter dependent on the DNA methyltransferases1/ Ubiquitin-like containing PHD Ring Finger 1 (DNMT1/UHRF1) complex, which in turn stimulated NF-κB activity [
119].
3.8. Slow growing cells
Tumour cells may have transcriptional plasticity, due to epigenetic reprogramming, which will change them into persister cells. These "persisters" are a collection of cells that are growing slowly and have the potential to either re-grow when therapy is stopped or develop enduring resistance. KDM5B, a member of the KDM5A family, designates a small subset of slow-cycling cells in melanomas that are necessary for ongoing tumour maintenance and are dynamically triggered depending on the microenvironmental situation. These KDM5B-positive cells cycle slowly and have increased self-renewal. They are intrinsically resistant to many cytotoxic therapies, and through a dysfunctional Jagged 1/Notch 1-signalling pathway, they can produce an offspring that are extremely proliferative [
120].
Recent research demonstrated that abnormal expression of nerve growth factor receptor (NGFR), SRY-Box transcription factor 2 (SOX2), AXL RTK and melanocyte inducing transcription factor (MITF) in melanoma cells make them more susceptible to shift into a persister state in response to RAF and MAPK inhibition [
121,
122].
In response to targeted kinase inhibitors, the histone H3 lysine 27 trimethylation (H3K27me3) specific demethylases, KDM6A/B, are activated and crucial for the transformation of naive glioblastoma stem cells into the slow-cycling drug-tolerant persisters (DTPs). Pervasive acetylation (H3K27ac) of cis-regulatory components occurs in conjunction with the transition to the persister state and is made possible by a widespread redistribution of the repressive mark H3K27me3. These persisting cells display primitive neurodevelopmental hallmarks because of this modified chromatin state and heavily rely on Notch signalling [
123]. Sharma et al. consistently identified a small fraction of reversibly "drug-tolerant" cells while simulating the acute response to several anticancer drugs in drug-sensitive human tumour cell lines. These cells exhibit a >100-fold decrease in drug sensitivity and continue to exist due to activation of the insulin-like growth factor 1 receptor (IGF-1R) signalling and a modified chromatin state that needs the histone demethylase RBP2/KDM5A/Jarid1A. Individual cells within the population transiently acquire this drug-tolerant phenotype at low frequency, suggesting that drug tolerance is dynamically regulated by phenotypic heterogeneity [
112]. In addition, KDM5A is necessary to create a transient chromatin state in EGFR-mutant lung cancer cell lines with elevated expression driven by IGF-1 signalling pathway in both DTPs and drug-tolerant expanded persisters (DTEPs). This will mediate the development of EGFR inhibitor resistance [
112].
The irreversible stop of cell growth known as cellular senescence is what causes tumour-suppressive pathways regulated by p16 and/or p53 to be activated. As a tumour suppressor, the protein p16
INK4a (also known as p16) inhibits the activity of cyclin-dependent kinases (CDKs) and slows cell division by delaying the transition from the G1 to the S phases of the cell cycle [
124]. Both endogenous and external factors can promote cellular senescence. The three main factors are shortening of telomere, increased mitogenic signalling created by oncogene activation, and non-telomeric DNA damage brought on by chemotherapeutic medicines. Senescence can begin, for instance, when chemotherapy drugs like doxorubicin and cisplatin cause cell death [
125,
126,
127]. In part via inhibiting apoptosis, p53 and INK4a/ARF mutations encourage carcinogenesis and treatment resistance [
126]. Drug resistance and tumour progression/recurrence have been linked to a mechanism known as escape from therapy-induced senescence (TIS) [
128]. The ability of cancer cells with TIS to acquire stem-cell characteristics explains how they can avoid senescence and relapse [
129,
130].
Moreover, metastasis, chemoresistance, and cancer recurrence are all influenced by tumour dormancy. CSCs frequently exist in a quiescent state where they might stay in the G0/G1 stage and proliferate at a slow rate [
131,
132]. Quiescence (reversible cell cycle arrest) features help CSCs develop resistance to radiation and chemotherapy because most traditional chemotherapeutic agents target proliferating cells [
87,
133,
134]. For example, the majority of 5-FU-resistant gastric cancer cells with CSC characteristics were quiescent cells that stayed in the G0/1 phase [
135]. In response to chemotherapies, CSCs enter quiescence by initiating a complex array of intracellular molecular and epigenetic programmes [
131]. The three signalling pathways most frequently engaged in CSC quiescence are Notch, Wnt, and p38-MAPK. Active p38 mitogen-activated protein kinase 1 (MAPK1) can cause CSC to enter a dormant state in prostate cancer [
136]. It is noteworthy that the Wnt canonical pathway component c-Myc can speed up the CSC cell cycle and encourage CSC reawakening, whereas their inactivation was directly linked to the onset of reversible quiescence [
137,
138,
139,
140].
3.9. Undruggable targets
Several of the most powerful oncogenes and tumour suppressor genes, such as MYC, RAS, and TP53, remain intractable despite increasing progress in efforts to target oncogenic driver mutations. Ras proteins were discovered to be oncogenes in the early 1980s, but despite extensive research over more than three decades to identify particular inhibitors, they were thought to be unreachable targets.
In up to 90% of human melanoma, mutated BRAF or mutated NRAS hyperactivate the kinase ERK, according to the examination of genetic changes [
141,
142]. The rationale for developing targeted inhibitors of mutant BRAF and MEK, the kinase that functions downstream of BRAF to activate ERK, as treatments for advanced melanoma was supplied by these findings [
143]. The overall survival of patients significantly increased as a result of the introduction of targeted medicines (MAPK pathway inhibitors such as BRAF and MEK inhibitors) and immunotherapies (immune checkpoint inhibitors). However, a lack of clinical effects, side effects, and the rapidly escalating treatment resistance limit the long-term efficacy of such treatments. This resistant phenotype is supported by several molecular pathways [
144]. Moreover, resistance may also be caused by target indifference, in which the effects of focusing on an oncogenic driver are mitigated by changes made to the pathway later on or concurrently. This is demonstrated by the fact that resistance to anti-EGFR therapy in colon cancer can be caused by downstream mutations that activate NRAS or KRAS [
145]. Recently it was shown that JAK/STAT pathway activation occurs as BRAFV600E thyroid cancer cells become resistant to BRAF inhibitors [
146]. Interestingly, insensitivity to inhibition of the MAPK/ERK pathway in advanced melanoma tumours harbouring the BRAFV600E mutation resulted from the activation of compensatory signalling cascades. Particularly in mesenchymal-like cells, the PI3K/Akt/mTOR axis displayed increased activity, resulting in a decreased MAPK/ERK signalling dependency and promoting stem-like features making the latter pathway's inhibitors ineffective [
147].
Cancer frequently harbours mutations in the p53 pathway. In fact, the TP53 gene exhibits mutations or deletions in around 50% of human malignancies, which predominantly cause decreased tumour suppressor activity [
148]. Damaged cells may multiply after losing their p53 functioning, passing on changes to the following generation [
149]. Deregulation of p53 frequently causes tumour development and mutant p53 cancers are frequently characterised by genomic instability promoting proliferation, migration, invasion, angiogenesis and increased drug resistance [
149,
150].
In NSCLC, mutated p53 increases binding to the nuclear factor erythroid 2 –related factor 2 (Nrf2) promoter, supported by an activation of the NF-κB signalling pathway, which further increases Nrf2 expression. Nrf2 is a transcription factor that codes for detoxification enzymes and confers resistance to anticancer drugs. In addition, in p53 mutant colon cancer cells, absence of DNA mismatch repair trigger resistance to cisplatin [
151]. Furthermore, mutant p53 affects the ERK-mediated transcription of early growth response-1 (Egr-1) and ERK pathway which enhance the production of EGFR ligands and stimulates EGFR signalling, rendering therapy to EGFR-inhibitor ineffective [
152]. Moreover, mutations in PI3K and MAPK pathways are common in metastatic CRC and accelerate tumour growth in conjunction with other prevalent mutations in the p53, TGF-β and Wnt signalling pathways [
153] . Mutations in the MAPK pathway are present in these CRC patients (0.8% in MAP2K1, 1.7% in MAP2K4, 3.9% in NRAS, 8.5% in BRAF and 44% in KRAS). The PI3K/Akt/mTOR pathway is mutated in CRC patients (1% in AKT1, 2.4% in PIK3R1, 2.5% in PIK3CG, 2.8% in PTEN and 18% in PIK3CA) [
5]. Additionally, 11% of the remaining patients exhibit mutations in RTKs which are upstream of both pathways triggering the emergence of resistance mechanisms to chemotherapy or targeted therapies [
15,
154].
Breast, colorectal, liver and other cancers are all mostly driven by the MYC oncogene. More than 70% of human malignancies exhibit high and/or abnormal Myc expression, which is associated with aggressive diseases and a bad prognosis [
155,
156]. Myc is a difficult oncoprotein to target due to its high frequency of overexpression in malignancies and its pervasive function in transcriptional control. There are presently no specific medications that can be used to target Myc, primarily due to its "undruggable" characteristics: Myc is primarily localised in the nucleus, making it inaccessible to antibodies and lacking an enzyme site where typical small molecules can bind [
157]. BRD4 is a crucial epigenetic regulator (a chromatin regulator) and a member of the BET family. The human genome contains regulatory components including silencers (repressors), enhancers/super-enhancers and promoters that are used to dynamically modulate the regulation of transcription. In BET inhibitor-sensitive leukaemia cells, the classic enhancer or super-enhancer controls MYC expression through BRD4 binding. The expression of MYC is inhibited and cell proliferation is suppressed as a result of BET inhibitor’s blocking of BRD4's ability to bind to its genomic targets. However, by various mechanisms, long-term drug therapy may restore MYC expression. One of those mechanism is maintaining MYC expression by activating Wnt/β-catenin signalling pathways which result in enhanced β-catenin binding to the sites that were initially occupied by BRD4 leading to drug resistance [
158].
The assessment of tumour heterogeneity is a crucial clinical concern. Genomic sequencing is being used to assess heterogeneity in cancer samples that were either archived at the time of diagnosis or later biopsied upon recurrence. This method has significant limitations because it is unlikely to adequately capture tumour heterogeneity, which has clear consequences for cancer therapy despite its utility in some circumstances for therapy selection [
159]. Targeting an 'actionable' driver mutation, for instance, might only be successful if the mutation is truncal (i.e., clonal and present in the majority of subclones and parts of the tumour during the course of its lifetime) [
160]. In other situations, the presence of a particular mutation may not indicate that it is clonal, and vice versa, the scarcity of a mutation does not indicate that it is accidental. In fact, resistance to targeted medications can be brought on by subclonal driver mutations in the PI3K pathway genes and ESR1. A list of the 'clonality' of driver mutations might be helpful in this case [
161,
162].
3.10. Tumour Microenvironment
Cancer cells, stromal cells, ECM, blood and lymphatic vessels, immune cells, nerve fibres, signalling molecules and related acellular components make up the TME. This latter is sculpted and instructed by cancer cells to support the emergence of cancer hallmarks, react to stimulation, internal or external stress and therapy, and eventually support the survival, growth, angiogenesis, migration, invasion, and immune evasion as well as drug resistance of these cells [
10].
TME consists of myeloid-derived suppressor cells (MDSCs), mast cells, CAFs, TAMs, vascular endothelial cells, adipocytes, pericytes, tumour-associated neutrophils, dendritic cells, and granulocytes. It also includes malignant cells, NK cells, T and B cells. Cancer is protected from immunological eradication by the suppressive immune microenvironment [
4,
163]. Regulatory T (Treg) cells, neutrophils, macrophages, MDSCs, CD4
+, FOXP3
+, and CD25
+ assist in establishing an immunosuppressive pre-metastatic microenvironment [
164,
165]. The activation of MDSCs, TAMs, and CAFs by reactive oxygen species (ROS) was demonstrated to be crucial in strengthening their immunosuppressive functions [
166,
167]. Immune cell recruitment into the TME can be affected by the ECM. For example, the ECM can activate the pro-survival pathway PI3K/Akt, which makes it easier for CSCs to evade the immune system [
168]. The recruitment of immunosuppressive cells like Tregs and TAMs by ECM proteins has also been demonstrated to support CSC survival while inhibiting the recruitment of cytotoxic T cells, which are anti-tumourigenic immune cells [
169,
170,
171]. Moreover, lipid metabolism has been associated with tumour progression, recurrence, and exhaustion of CD8 T cell through activation of programmed-cell death protein-1 (PD-1), which results in escaping the immune surveillance after treatment [
172,
173].
Key aspects that define cancer stemness, the recruitment of non-malignant cells that support tumour cells and ECM remodelling are coordinated by cellular crosstalk via several signalling network such as juxtracrine and paracrine pathways [
174]. The suppression or modification of interferon-gamma (IFN-γ) signalling, activation of the MAPK and Wnt/β-catenin pathways, a decreased T-cell response and tumour antigen production are a few often found pathways that inhibit the immunotherapy response leading to treatment resistance [
175].
Avoiding detection and eradication by the immune system results in multidrug resistance [
176]. PD-1 are frequently expressed on the membranes of immune cells such as macrophages, T and B cells. While various tumour cells express programmed death ligand 1 (PD-L1). It has been demonstrated that the interaction of PD-1 and PD-L1 on T cell surfaces can inhibit the activity of killer T cells by promoting apoptosis, which causes tumour cells to escape the immune system [
177]. Through the IL-6/STAT3/PD-L1 axis, CAFs modulated neutrophil activation, survival, and function in tumour tissues in HCC to promote immune suppression [
178].
MSCs can produce a wide range of cells that engage in paracrine signalling, including IL-6 and IL-8, advancing the development of cancer and enhancing chemoresistance [
179]. When exposed to cisplatin, instead of going through apoptosis, a subpopulation of cisplatin-resistant MSCs activate a phenotype linked to senescence [
179]. As a result, various proteins (such as PLC-y1, RSK1/2/3, WNK1, c-Jun and p53) get phosphorylated, activating signalling pathways resulting in secretion of IL-6 and IL-8 into the TME. When breast cancer cells and MSCs were co-cultured simultaneously, the therapeutic impact was diminished in vivo due to the upregulation of resistance-related genes (such as MUC1, MYC and BRCA1) in the breast cancer cells after cisplatin pre-treatment [
179].
MSCs can differentiate into CAFs. Recently, it was revealed that CAF triggered TKI resistance in HCC via the activation of PI3K/Akt/mTOR and RAF/ERK/STAT3 pathways [
180]. Moreover, it was determined that the major signalling pathway activated by CAF is STAT3 driving everolimus resistance in neuroendocrine tumours cells [
181].
In oesophageal squamous cell carcinoma, PAI-1 secreted by CAF activate the MAPK and Akt pathways in a paracrine manner resulting in production of ROS, induction of DNA damage and cell death leading to chemoresistance [
182]. Additionally, drug resistance was promoted in tumour cells via NF-κB pathway induction by CAF-derived paracrine signals, such as exosomes, metabolites and chemoattractant cytokines [
183,
184]. CAFs can also enhance stemness through NF-κB signalling activation in gastric cancer [
185]. Moreover, CAF enhanced the stemness of HCC by activating the Notch1 signalling pathway [
186]. Furthermore, recently it was discovered INF-γ/STAT1/Notch3 as a molecular connection between CSCs and CAF using a bioinformatics strategy in TNBC cell lines resistant to doxorubicin [
187].
The cellular composition and functional state of the TME will differ depending on the organ in which the tumour is located, as well as on cancer type and stage which will affect the delivery of treatment leading to a heterogeneous exposure to anticancer drugs [
13,
188,
189]. TME can be divided into six different types of specialised microenvironments: the hypoxic, immunological, innervated, metabolic, mechanical and acidity niches. All these niches interact together and facilitate the progression and drug resistance of cancer [
190].
Depending on their location within the cancer tissue, the cells in the tumour mass grow in a 3D tissue structure and are exposed to oxygen unevenly. As opposed to the tumour core, which is poorly vascularized, blood vessels in tumour tissues are typically randomly arranged and only cover the outer portion of the tumour mass [
189]. A hypoxic microenvironment is created within the tumour core as a result of increased tumour cell proliferation, which places the cells there further away from the supporting blood vessels than the cells outside the tumour. This can result in varied treatment responses. By increasing the expression of genes linked to cell survival, angiogenesis, and anti-apoptotic pathways, tumour cells respond to hypoxic circumstances and the modified TME leading to the progression of cancer and the development of treatment resistance [
191,
192,
193].
Interestingly, cancer cells may proliferate and colonise in anatomical areas which are sanctuary sites where medications administered systemically are unable to reach the therapeutic window. The brain's blood-brain barrier (BBB) and the central nervous system (CNS) are the two most typical examples [
194]. Additionally, the peritoneum is another sanctuary site in severe paediatric leukaemia that may be treated with intra-peritoneal chemotherapy and tests that result in the management of preventative emission. Among these sanctuaries, the CNS is conceivably the most resentful therapeutic necessity. The extent of CNS tropism is higher in some types of diseases including melanoma, lung, breast, and kidney cancers. Those sanctuaries are physical barriers that lead to devastating clinical outcome [
195].
The TME causes chemotherapeutic resistance via intrinsic or acquired mechanisms. Cancer dormancy, stemness and progression as well as intercellular communication, redox adaptability and drug resistance are reprogrammed by hypoxia [
196]. Hypoxia affects the TME and treatment efficacy by encouraging cancer cells greater production of hypoxia-inducing factors (HIFs), most frequently HIF-1α. This latter stimulates the transcription of numerous genes, including vascular endothelial growth factor (VEGF) which enhance angiogenesis and as a result, cancer cells are better able to sustain their oxygen supply and metabolism, improving their chances of surviving [
197,
198]. Increased somatic mutational burden of oncogenes and tumour suppressors, such as TP53, MYC and PTEN is also linked to the hypoxic niche [
199]. Cancer cells with p53 mutations or suppressed p53 transcription have the ability to avoid p53-mediated apoptosis pathways under hypoxic conditions, leading to the selection of cancer cell clones and the production of apoptosis-resistant cells [
200]. Under hypoxic conditions, it has been demonstrated that p53 transcriptional activity is inhibited and the expression of efflux pumps, ABCB1 and ABCB5 is increased once HIF-1α binds to p53 in ovarian cancer cells, promoting their resistance to commonly used chemotherapeutics [
201].
One of the characteristics of cancer is metabolic reprogramming, which is a modification in metabolism or nutrition supply. Increased metabolism of glutamine, glucose, amino acids, lipids, addiction to ROS and accumulation of lactate are common characteristics of cancer [
202,
203,
204]. The synthesis of brain-derived neurotrophic factor by CAFs was driven by lactate in cancer cells in an NF-κB -dependent way, which in turn activated TrkB/Nrf2 signalling in cancer cells to lessen their susceptibility to anlotinib [
205]. These results support the connection between drug resistance, metabolism, and NF-κB signalling.
Cancer is characterized by dysregulated pH, which is one of the TME variables. Extracellular pH (pHe 7.3–7.5) is often higher than intracellular pH (pHi 6.8–7.2) in healthy tissues and cells, while cancer cells generate a "reversed pH gradient" with increased internal pH and decreased external pH [
206,
207,
208]. This reversed pH gradient makes it difficult for cancer cells to undergo apoptosis and prevents them from dying off [
209,
210]. Cancer cells' acidic extracellular environment (pH 6.5–7.1) plays a role in their chemotherapy resistance [
211]. Recent research showed that an acidic tumour environment promotes cellular stemness and increases radio- and chemoresistance in oral cancer cells by causing increased cancer cell migration [
212]. Acidic environments are extremely stressful for cells triggering many signalling pathways and likely activating powerful survival signalling pathways, such as those linked to cell stemness and undifferentiation leading to an increase in treatment resistance. Melanoma, neuroblastoma and breast cancer cells become more invasive and undergo an increase in oxidative phosphorylation and EMT in an acidic niche [
213,
214,
215]. The development of acidic niches is also influenced by the activation of oncogenes like Ras and Myc and the inactivation of tumour suppressors like p53. Acidic pHe produces resistance to daunorubicin by inducing the activation of P-gp and the subsequent activation of p38 MAPK [
216,
217]. Inhibition of apoptosis in colon cancer cells is also associated with tumour acidity and p53 function loss [
218]. Moreover, the absorption and resistance to cisplatin in melanoma cells are influenced by an acidified TME [
219].
Neurology and cancer science are closely related, with neurotransmitters and neuropeptides generated from the nerve creating an "innervated niche" [
220,
221]. The neuroligin-3 (NLGN3)-stimulated PI3K/mTOR pathway, which is activated by active neurons, aids in the formation of high-grade gliomas [
222]. Paracrine stimulations of cGAMP to astrocytes, cytokines production, activation of STING pathway and NF-κB and STAT1 signalling are triggered in brain metastatic cells via gap junctions between astrocytes and lung/breast cancer, which promotes cancer growth and resistance to chemotherapy [
223,
224].
The creation of a mechanical niche depends on stromal cells, extracellular and intracellular components, and intercellular signalling [
225]. There are various structural proteins in the ECM such as collagen, laminins, fibronectin, elastin, proteoglycans and glycoproteins. The ECM is a 3D network of macromolecules that provides the biochemical and biophysical characteristics of the non-cellular bulk surrounding the cells. Additionally, non-malignant tumour-associated stroma cells are a crucial component of the TME, altering tumour characteristics, illness prognosis, and therapeutic response. Cell surface proteoglycans, cell adhesion molecules such as integrins, and hyaluronic acid receptors such as CD44, mediate biochemical and biophysical signalling as well as cell anchoring to the ECM [
189,
226]. For instance, in breast cancer increased laminin-mediated signalling and overexpression have been connected to diminished treatment responsiveness and improved tumour cell invasion and metastasis [
227]. Fibronectin-integrin β1 interactions activate the PI3K/Akt and MAPK/ERK 1/2 pathways leading to chemotherapy resistance [
228]. The integrin β1 downstream kinases FAK and Src are activated in HER2+ breast cancer cells that are resistant to lapatinib (a HER2-targeted therapy), resulting in these overcoming HER2 inhibition [
229].
Matrix cells in the TME communicate with cancer cells through exosomes. Exosomes are small, bilayered molecules involved in autocrine, paracrine and endocrine signalling that are released by stromal and cancer cells in the TME. Altering vital survival signal transduction pathways, inducing EMT, activating anti-apoptotic pathways, and modifying the immune system are just a few of the ways that exosomes can make tumour cells resistant to treatment [
230]. Exosome-mediated transfer of different ncRNAs, such as lncRNAs and miRNAs, may be a way for cancer cells to develop treatment resistance by causing genetic and epigenetic changes [
230,
231]. Recently it was shown that miR-1228-3p carried by CAF-derived extracellular vesicles increases HCC's chemoresistance by activating the PI3K/Akt pathway [
232]. It was also revealed that Wnt/β-catenin and BMP signalling diminish the susceptibility of hepatoma cells to sorafenib and promote EMT in CAFs-derived Gremlin-1-rich exosomes [
233]. Moreover, it was demonstrated that CAF-derived exosomes harbouring miR-20a can encourage chemoresistance and aggressive growth in NSCLC cells via the PTEN/PI3K/Akt signalling pathway [
234]. Exosomal miR-21 and IL-6 produced from CAFs together increased MDSC formation in oesophageal squamous cell carcinoma by activating STAT3, which made tumour cells resistant to cisplatin [
235]. Furthermore, SOX2 and PD-L1 expression was mediated by PI3K/Akt signalling pathway activation and showed to be a mechanism by which exosomes from CRC/MDR cells may increase cetuximab resistance in KRAS wild type cells [
236].
3.11. Epithelial-Mesenchymal Transition
The phenotypic change from epithelial to mesenchymal cells, or epithelial-mesenchymal transition (EMT), occurs when epithelial cells lose their cell identity and take on mesenchymal traits altering the cell's shape and expression of surface markers in the process [
237]. Epithelial cells, in the EMT process, experience depolarization, lose their cell-cell contact, adherent property, and develop elongated fibroblast-like morphology, is known to be triggered by ncRNAs, growth factors, cytokines, and hypoxia. These occurrences are accompanied by a concurrent increase in mesenchymal markers (integrin, laminin 5, N-cadherin, fibronectin, vimentin and type I collagen) and a concurrent decrease in epithelial markers (laminin 1, desmoplakin, E-cadherin and type IV collagen) expression. EMT is typically seen under healthy conditions, but tumour cells have the ability to carry out the same process while cancer is developing. Recent evidence suggests that pathological hyperactivated EMT is closely linked to elevated therapeutic resistance of cancer cells. Intracellular regulatory miRNA, exogenous inducers, epigenetic modulators, and cellular signalling pathways such as SMADs, PI3K, MAPK, ERK, TGF-β, Notch and Wnt/β-catenin are only a few of the molecular players involved in the regulation of EMT [
238,
239]. For instance, through the Wnt/β-catenin pathway, tongue squamous cell carcinoma cells gained cisplatin resistance and stem cell-like properties resulting in an enhanced EMT [
240]. Moreover, in oral cancer, Notch signalling increases the population of CSCs, improves angiogenesis and EMT, and responds strongly to the DNA damage response induced by cisplatin [
241]. TGF-β is the primary substance released by CAFs; it causes EMT and encourages the acquisition of gastric CSC features both of which eventually result in drug resistance [
242]. Furthermore, miR-155 is overexpressed in oral squamous cell carcinoma, which results in resistance to cisplatin by inhibiting the expression of FOXO3a and promoting the EMT pathway [
243].
Recently, it was demonstrated that cancer cells treated with chemotherapy release IL-1β triggering the release of integrin-αvβ1 and matrix metalloproteinase 9 causing the activation of TGF-β which in turn promote EMT in breast cancer cells [
244]. Moreover, family with sequence similarity 46, member A (FAM46A) activated TGF-β pathways promoting chemoresistance in ovarian cancer cells [
245]. TGF-β signalling promoted EMT and resistance to doxorubicin in breast cancer cells by upregulating lncRNA urothelial carcinoma-associated 1(lncRNA UCA1) [
246]. HIF-1α/TGF-β2/GLI2 signalling is responsible for chemoresistance in CRC cells [
247].
It was shown that hexokinase domain containing protein-1 (HKDC1) is essential for gastric cancer cell glycolysis, carcinogenesis, and EMT by activating the NF-κB pathway resulting in resistance to 5-FU, oxaliplatin and cisplatin in gastric cancer patients [
248]. For instance, epithelial ovarian cancer (EOC) cells resistant to paclitaxel, cisplatin, erlotinib and carboplatin displayed high NF-κB activity [
249]. The Notch signalling pathway is upregulated in breast cancer patients that are resistant to tamoxifen which can promote CSCs and EMT [
250]. Furthermore, as a result of the activation of PI3K/Akt/mTOR signalling, the expression of EMT and CSC markers was considerably increased in cisplatin-resistant EOC cells [
251].