Summary:

Cancer is characterized by loss of the regulatory mechanisms that preserve homeostasis in multicellular organisms, such as controlled proliferation, cell–cell adhesion, and tissue differentiation. The breakdown of multicellularity rules is accompanied by activation of “selfish,” unicellular-like life features, which are linked to the increased adaptability to environmental changes displayed by cancer cells. Mechanisms of stress response, resembling those observed in unicellular organisms, are actively exploited by mammalian cancer cells to boost genetic diversity and increase chances of survival under unfavorable conditions, such as lack of oxygen/nutrients or exposure to drugs. Unicellular organisms under stressful conditions (e.g., antibiotic treatment) stop replicating or slowly divide and transiently increase their mutation rates to foster diversity, a process known as adaptive mutability. Analogously, tumor cells exposed to drugs enter a persister phenotype and can reduce DNA replication fidelity, which in turn fosters genetic diversity. The implications of adaptive evolution are of relevance to understand resistance to anticancer therapies.

Complex multicellular organisms, including mammals, exploit sophisticated and tightly regulated networks and genetic constraints that subject the fitness of individual cells to the fitness of the whole organism. The evolution of multicellularity features underscores selection for cooperation between unicellular entities. Cells must proliferate only when required to fulfill their function, and cell death by apoptosis is sometimes essential to maintain tissue homeostasis (1). However, harmful events, such as somatic mutations or viral infections, may subvert multicellularity constraints, and the hallmarks of cancer (the common traits of human tumors) include many features reminiscent of a “selfish,” unicellular-like life (2).

During tumorigenesis, cancer cells forgo the cellular cooperation and the regulatory mechanisms that preserve homeostasis in multicellular organisms (3). Cancer cells display sustained proliferation, evade signals of growth suppression, resist programmed cell death, and redirect allocation of resources by promoting neoangiogenesis. Moreover, tumor progression is also characterized by progressive cellular dedifferentiation (ref. 2; Fig. 1).

Figure 1.

The hallmarks of tumor cells and the biological features of unicellular organisms. Cancer cells display properties that parallel the behavior of unicellular organisms. The biological, genetic, and metabolic features shared between cancer and bacterial cell populations include competition between clones, glycolytic metabolism, formation of communities by manipulating the environment, stress responses leading to increased genetic instability, and adaptation to hostile conditions such as drug treatments.

Figure 1.

The hallmarks of tumor cells and the biological features of unicellular organisms. Cancer cells display properties that parallel the behavior of unicellular organisms. The biological, genetic, and metabolic features shared between cancer and bacterial cell populations include competition between clones, glycolytic metabolism, formation of communities by manipulating the environment, stress responses leading to increased genetic instability, and adaptation to hostile conditions such as drug treatments.

Close modal

By dysregulating cellular processes associated with the transition from unicellular to multicellular life, cancer cells activate survival strategies, including rapid proliferation and adaptability to stressful environments, that had been perfected by autonomous organisms such as bacteria (4–7). In other words, cancer, a disease of multicellular organisms, shares biological features with prokaryotic cells (Fig. 1).

Keeping in mind the remarkable evolutionary distance between bacteria and mammalian cells, here we discuss the scientific and therapeutic implications of considering cancer cells as selfish forms of life which subvert multicellularity laws and behave as single competing units much like bacteria (7, 8).

Cancer can be considered as an atavistic form of life, characterized by competition among individual cells for access to nutrients, survival, and proliferation, similar to what occurs in bacterial populations (1, 7). As a consequence, several hallmarks and acquired capabilities of cancer cells are strikingly similar to properties displayed by microorganisms (2, 4). For instance, from a metabolic point of view, cancer cells mainly rely on anaerobic glycolysis (the so-called Warburg effect), which is the prevalent energy source among bacteria (9).

In natural environments, microorganisms face a constant battle for nutrients and have evolved several mechanisms to compete with other individuals for resources, including rapid growth, increased matrix production to secure favorable environments, and production of antimicrobial toxins (10, 11). Similarly, human cancers display high levels of molecular heterogeneity, and within a single lesion distinct subclones can compete with each other (12). This can be observed especially under treatment with anticancer agents, when previously unfit drug-resistant subclones outcompete the dominant drug-sensitive cells (13, 14).

Intriguingly, bacteria can also thrive as complex communities regulated by cooperation or competition, either between individuals from the same species or between distinct bacterial strains (4). For example, especially (but not exclusively) when facing hostile environments, bacteria can form biofilms, multicellular aggregates embedded in a proteinaceous extracellular matrix (15). The formation of a biofilm promotes survival of the population, as these complex structures have been shown to protect bacteria from antibiotics, toxins, and solvents (16).

Analogously, the interactions between cancer cells and their stroma can be compared with a special form of biofilm, whereby cancer cells orchestrate neoangiogenesis, recruitment of cancer-associated fibroblasts and immune cells to promote the survival of the entire community (2, 17, 18). Cooperation through paracrine signaling can also promote increased resistance to targeted treatment; for example, we previously showed that in colorectal cancer, treatment with anti-EGFR targeted therapy promotes increased secretion of TGFα and amphiregulin from resistant cells, which in turn protect surrounding sensitive cells in the population from EGFR blockade (19).

Both bacteria and cancer can deploy complex responses to hostile environments to protect the population from eradication. Some of the bacteria inside a biofilm are able to switch to planktonic state and colonize distant sites (3, 20). Moreover, under stressful conditions some individuals can undergo sporulation, generating highly resistant spores that germinate once favorable growth conditions are restored (15, 21). Similarly, some cells within a tumor (possibly in response to challenging circumstances) acquire metastatic competence, i.e., the ability to separate from the cancer of origin and colonize distant organs (22). At the site of colonization, disseminated tumor cells encounter an unfavorable environment, in which they enter a state of dormancy (just like spores) and slowly condition the surrounding stroma until a supportive niche is formed (23).

Notably, competition can lead to selection of variants that are better suited to colonize the environment. The evolution of tumors cells and asexual bacteria is governed by the interplay of genetic drift, heritable variation, and Darwinian selection (24). Experimental and computational evidence shows that constitutive mutators are present at low frequency in a bacterial population, because they result in the overproduction of lethal mutations. However, in stressful conditions (such as antibiotic treatment or limited access to nutrients), hypermutators are positively selected and can become fixed in the majority of the population (25–29). Similarly, genome instability is a hallmark of cancer and is favorably selected in tumor cells for its role in fostering the emergence of progressively fitter subclones and supporting tumor evolution (2).

The atavistic hypothesis states that tumorigenesis involves the reactivation of survival programs dating back to the evolution of unicellular organisms, and the concomitant disruption of complex features evolved to support the intercellular communications required for multicellular organisms' development and physiology (7, 30).

Transcriptional analysis of more than 3,000 tumor samples across seven different tumor types unveiled that cancers rely on unicellular processes for survival. Dysregulation of genes and cellular processes unique to multicellularity was detected across multiple tumor types, together with strong upregulation of genes conserved in unicellular organisms, including those involved in proliferative signaling, cell death escape, and genomic instability (5, 31). Expression of highly conserved genes is linked to drug resistance in cancer cells (32), and tumors often activate transcriptional programs associated with dedifferentiation (33, 34).

Analysis of the evolutionary age of genes involved in human cancer revealed increased mutational processes in genes younger than 500 million years, suggesting selection for somatic mutations occurring in younger genes preferentially linked to multicellularity (35).

Goode and colleagues used a computational approach to analyze different types of tumors from more than 9,000 patients, to unveil accumulation of point mutations and copy-number variations (CNV) in genes dating back to metazoan ancestors' genes. Their analysis showed that point mutations disrupt key regulators of multicellularity networks, and that CNVs dysregulate downstream effectors of these pathways (36).

Relatedly, genes with regulatory function in the communication between genes of unicellular and multicellular origin represent vulnerabilities that, when compromised, promote the emergence of tumorigenesis. For example, TP53, NF1, and PI3KCA genes, which are frequently mutated across different tumor types, act as regulatory hubs of multiple processes fundamental for the maintenance of the integrity of genomic networks (31, 36).

Whether atavism plays a role in cancer initiation and progression is still debated, as acquisition of cancer's biological features during tumorigenesis can be explained by stochastic genetic alterations and progressive strong environmental selection rather than reactivation of a conserved survival program. However, as discussed below, the two theories are not necessarily mutually exclusive and can coexist (37). According to basic laws of evolution, an enriched fraction of mutants in a population would increase chances of survival of a few individuals under selective conditions. Indeed, unicellular organisms deploy strategies of adaptive mutability to foster evolution in strongly selective environments (see below). Intriguingly, we and others recently observed that cancer adaptation to stress is at least initially fostered by activation of similar “atavistic” survival programs observed in bacteria under stress, thereby promoting mutagenesis and genetic diversity; selection and expansion of phenotypes capable of replicating under stressful environments then occurs (refs. 38, 39; see details below).

When facing stressful environments, bacteria deploy rapid and reversible survival programs that foster genetic diversity and facilitate adaptation to changes in the environment (5, 40, 41). Effective strategies to evade a stressful condition include relocating to a new environment through swimming and the formation of biofilms, as discussed above (3, 4).

An additional fascinating strategy exploited by unicellular cells to evade stress-induced death relies on temporary phenotypic switches. Indeed, in bacteria a fraction of cells named “persisters” evades antibiotic killing by entering a physiologically dormant state displaying transient phenotypic stress tolerance, without undergoing genetic changes (42, 43). Persister cells, different from permanent genetically resistant cells, reinitiate growth and regenerate a drug-sensitive population upon termination of drug treatment. Importantly, development of resistance eventually occurs upon prolonged drug treatment of persister cells, representing a major cause of antibiotic failure, which leads to recurrent bacterial infections (43).

Many of these stress adaptations are tailored to the specific stress. For example, the SOS DNA damage response induces activation of error-prone DNA polymerases that can bypass the damage and terminate replication, while in case of high temperatures, induction of heat-shock chaperones allows the resolution of misfolded or aggregated proteins (44).

The SOS response is triggered by DNA damage caused by cell malfunctioning, stalled DNA replication forks, or conditions such as oxidative stress and antibiotic treatment. In E. coli and related bacteria, the coating of RecA protein to single-strand DNA (ssDNA), generated either as a result of DNA damage itself, following stalled DNA replication forks, or during the DNA-repair process, induces the autoproteolytic cleavage of LexA, a transcriptional repressor, leading to upregulation of SOS regulon genes (45, 46). The SOS regulon includes genes involved in multiple functions, such as DNA recombination and repair, nucleotide excision repair, DNA synthesis past damaged bases, and control of cell division (45, 46). Among these, the activation of specialized low-fidelity DNA polymerases, which temporarily replace canonical replicative polymerases, is responsible for a mutagenic form of DNA replication associated with increased rate of base substitutions and indels (47–49).

However, in addition to stress-specific responses, many bacteria deploy a strong general stress response to react to a variety of growth-limiting conditions and stresses, such as nutrient deprivation, stationary phase growth, DNA damage, and extreme temperatures or pH (44, 50). In E. coli and related bacteria, the master regulator of the general stress response is the sigma factor RpoS. Different mechanisms regulate the increase of RpoS levels and the induction of the multiple RpoS-dependent effectors; indeed, some are dependent only on RpoS, whereas others require certain conditions and additional inputs (44, 50).

Both RpoS and SOS responses seem to play a role in persister formation. In particular the SOS-dependent DNA-repair functions are central for persister formation and recovery. Indeed, emergence of surviving persisters in response to DNA-damaging agents is impaired in SOS-deficient mutants (43).

Cancer cells are constantly exposed to intracellular and extracellular stressful stimuli, such as hypoxia, reduced nutrient availability, glucose deprivation, and DNA damage. In order to foster adaptation and survival, cells deploy a variety of stress responses to deal with adverse environments. Indeed, cancer cells activate multiple survival strategies, alter gene expression, reprogram metabolic pathways, and trigger stress-response mechanisms that can promote mutability and genetic diversity (5).

Cancer cells, like bacteria in response to antibiotics, can enter a reversible drug-tolerant persister state when exposed to targeted therapies (51, 52). Through nongenetic mechanisms of drug tolerance, slowly replicating cancer persister cells escape death and avoid tumor eradication (53). Of clinical relevance, extended treatment of persister cells inevitably leads to the development of resistance and consequent treatment failure (52, 54).

In mammals, the mechanistic target of rapamycin kinase (MTOR) has been proposed as a possible analogue of the bacterial RpoS general stress regulator, which controls the switch to mutagenic break repair in E. coli (5). MTOR has a central role in sensing environmental conditions, including lack of growth factors, nutrient deprivation, low oxygen, and DNA damage, and regulates many fundamental cell processes, such as protein synthesis, metabolism, cell growth, autophagy, and aging (55). Dysregulation of mTOR regulatory signaling is implicated in cancer progression, and mTOR inhibitors have been developed as anticancer agents (55).

Cells are continuously exposed to internal and external sources of damage and have therefore evolved an intricate network of signaling pathways and repair mechanisms to cope with DNA lesions and protect genome stability. Interestingly, several error-prone DNA polymerases have also been described in eukaryotic cells, including yeasts and human cells (49).

Why are error-prone DNA polymerases conserved in highly developed organisms, if programmed cell death allows the elimination of cells with damaged genomes?

In human cells exposed to nonstressful conditions, when the replication fork stalls upon encountering a site of DNA damage, canonical replicative DNA polymerases are replaced by specialized polymerases characterized by poor accuracy in nucleotide incorporation and increased rates of base substitutions and frameshifts, ranging from 10−3 to 10−1 errors per base pair (49, 56); propensity to incorporate nucleotides using aberrant DNA primer ends; absence of proofreading 3′→5′ exonuclease activity; and moderate-to-low processivity (49, 57).

The expression of error-prone DNA polymerases is tightly regulated. In physiologic conditions, they are involved in multiple biological functions, such as nonhomologous end joining (NHEJ) and translesion synthesis (TLS) DNA-repair processes (58), somatic hypermutation of immunoglobulin genes (59), and protection against UV-induced genetic instability and skin cancer, through the prevention of replication fork collapse and double-strand breaks (DSB) accumulation (60).

However, according to mathematical models, in growth-limiting hostile environments the evolutionary costs of high-fidelity DNA replication may exceed the costs of error-prone replication, becoming therefore a counterproductive strategy, whereas the proliferation and survival of cells with limited DNA-repair capacity is favored, both in microbes and in cancer cells (61). This suggests that in some instances, also in multicellular organisms, cell survival is prioritized over genome perfection and occurs by downregulation/repression of DNA-repair mechanisms, which is preferred over stalling of DNA replication.

Indeed, DNA damage tolerance (DDT) response, evolved side by side with DNA-repair mechanisms, functions in circumstances where the possibility to make errors has a selective advantage. Through the involvement of specialized polymerases able to bypass DNA damage, DDT allows the replication of a damaged template, and postpones the repair at later time points, thus lowering the overall risk of replication fork collapse, genome instability, genetic rearrangements, and cell death (58).

The association of DDT with cancer is double-faced. On the one hand, defects in DDT are associated with cancer susceptibility and aging (58). On the other hand, uncontrolled overexpression of error-prone polymerases has been associated with accumulation of mutation and tumorigenesis in lung tumors (62), poorer prognosis in patients with glioma (63), and drug resistance (64). Notably, the involvement of DDT and error-prone DNA polymerases in stress responses affects the mutational profile, leading to a temporary increase of genetic diversity, which can favor adaptation to a hostile environment and foster evolution (38, 39, 48).

Mutations represent the primary source of genetic variation and, together with environmental selection, are an important evolutive driving force, in both cancer cells and bacteria (65).

Growing evidence shows that multiple microorganisms, such as bacteria and yeast, adaptively combine stress responses that transiently increase their genomic instability, even during replicative quiescent state, in response to different environmental stresses (5, 41, 66). The resulting stress-induced adaptive mutability promotes stochastic emergence of fitter mutants, leading to higher rates of adaptive evolution (41, 45, 66).

Bacteria enter a dormant state when life is unsustainable and recover when it is beneficial to do so. Acquisition of mutations during the stationary phase indicates that cells exposed to stress respond by promoting mutability; a subset of the newly acquired mutations fosters adaptation to the stressful environment.

Indeed, in a constant environment, well-adapted populations evolve with a constant low mutation rate, while, when organisms are maladapted to their environments (e.g., in conditions of starvation or when exposed to hypoxia or antibiotic treatment), hypermutator phenotypes can emerge (66, 67). These phenotypes transiently provide elevated mutation rates when populations experience bursts of stresses. However, when a new (adapted) population emerges, the mutator phenotype is no longer an advantage and is counterselected, and eventually the genome of the expanding adapted mutants returns to low mutation rates (45, 66).

In E. coli, stress-induced mutagenesis via mutagenic DNA break repair involves three components: (i) repair of DSBs; (ii) activation of the SOS damage response, which upregulates error-prone DNA Pol IV; and (iii) activation of the RpoS-mediated general stress response, which allows the accumulation of Pol IV errors in acts of DSB repair (5, 45, 66). This stress-induced adaptive mutability appears to induce mutagenesis in proximity to DSBs, creating clusters that reduce in density with distance from the DSBs (68). Stress-induced mutagenicity generates distinct forms of genetic changes, including single-nucleotide variants, insertions and deletions (indels), CNVs, and chromosomal rearrangements (45, 66). Induction of error-prone DNA polymerases and other genes of the SOS regulon is required for most point mutagenesis.

Permanent loss of highly conserved mismatch-repair (MMR) proteins, responsible for the recognition and repair of mispaired bases and 1-base insertion/deletion, generates constitutive mutators (69). Interestingly, the function of the MMR system is temporarily disabled during adaptive mutability in response to stress. Indeed, overexpression of one or more MMR proteins reduces stress-induced mutations (66).

Downregulation of DNA-repair capacity, associated with overexpression of error-prone DNA polymerases, increases the number of errors that persist and become mutations, therefore translating into more mutagenic replication of DNA and genomic instability (70).

Notably, it has been shown that even sublethal concentrations of antibiotics induce downregulation of the MMR system and upregulation of error-prone DNA polymerases which, in turn, promote increased genetic instability (70). This temporary increase in mutation rate promotes the generation of mutants and ultimately the acquisition of resistance to a wide spectrum of drugs, including compounds unrelated to the antibiotic applied (71).

Although it is easy to conceive that adaptive mutability is essential for the survival of unicellular organisms exposed to stress, multicellular organisms evolved multiple constraints to ensure genetic integrity and homeostasis, such as the tightly controlled recruitment of error-prone DNA polymerases in the replication of damaged DNA templates described above (5).

However, it has been shown that, when exposed to stressful conditions, tumor cells exploit survival strategies resembling bacterial stress-induced mutagenesis (38, 39). For example, constitutive activation of the TGFb axis causes downregulation of homologous recombination (HR)–mediated DSB repair, increased genetic instability, and clonal diversity, thus boosting the ability of cancer cells to adapt to drug treatment and the development of chemoresistance (72). Analogously, cancer cells exposed to hypoxia activate a transcriptional regulatory response that downmodulates DNA-repair mechanisms such as MMR and HR (73). Moreover, hypoxia promotes upregulation of error-prone DNA-repair pathways, such as NHEJ, and the use of TLS DNA polymerases (73). This switch from highly accurate to error-prone DNA repair creates a permissive milieu for genomic instability; in fact, hypoxic cancer cells display increased mutability and accumulation of frameshift mutations at repetitive microsatellite sequences in reporter plasmids (74).

The remarkable capacity of tumor cells to adaptively modulate their mutagenicity has also been observed in response to targeted agents, similar to bacteria in response to antibiotics.

Indeed, we recently showed that colorectal cancer cells, under stress imposed by targeted therapy, activate a response recapitulating key element of bacterial stress-induced adaptive mutability. Treatment with the anti-EGFR monoclonal antibody cetuximab, alone or in combination with the BRAF inhibitor dabrafenib, induced transient deficiency of both MMR and HR-mediated DNA-repair proficiency, and a concomitant switch from a high- to a low-fidelity DNA replication (38). Notably, treatment with these non–directly genotoxic drugs caused increased DNA damage and the observed response, in turn, translated into increased genetic instability. The stress-induced adaptive mutability phenotype in cancer cells, as in bacteria, was transitory and confined to maladapted cell populations. When the stress was terminated, either by drug withdrawal or emergence of permanently resistant cells, the MMR and HR levels reverted back (38).

We propose that during tumor development, when breakdown of multicellularity occurs, a stress response aimed at promoting survival over functional and genetic integrity is expediently restored.

Adaptive mutability in response to stress appears to be an evolutionarily conserved process rather than a phenotype restricted to colorectal cancers. Indeed, Cipponi and colleagues observed induction of DNA damage and a switch to mutagenic DNA replication process in melanoma and prostate, breast, and pancreatic cancers exposed to nongenotoxic drugs. This in turn resulted in genetic diversity and development of secondary resistance (39).

Notably, both in melanoma and colorectal cancer cells, the modulation in DNA-repair machinery was not simply a consequence of DNA damage or cell-cycle arrest, but was exquisitely linked to the inhibition of the specific oncogene addiction across tumor types, and was abolished when targeted treatment was applied to resistant cells (38, 64).

New mutations are constantly generated and fixed in cancer cells at a rate that, unless influenced by environmental agents or defects in the mechanisms of DNA repair, in many cases reflects the mutation rate of the tissue of origin (75). However, during cancer development and progression, cancer cells are constantly exposed to multiple stresses and changing environments (such as hypoxia, nutrient deprivation, metastatization to different organs, and ultimately therapy), which can affect their mutagenic ability (74). These punctuated bursts of evolution increase the probability that genetic alterations conferring survival advantage in the environment might be acquired (14). Similarly, targeted therapies represent yet another selective condition where increased generation of variants could facilitate the evolution of fitter subclones and ultimately the survival of cancer cells.

Although the role of stress-induced adaptive mutability in the acquisition of resistance to targeted therapies in cancer requires further elucidation, several indications point in this direction. We observed treatment-induced loss of MMR protein expression in clinical specimens obtained at maximal tumor response (nadir) in patients with MMR-proficient colorectal cancer treated with targeted therapies (38). Moreover, increased mutability at microsatellite regions was detected in preclinical models of acquired resistance (38).

Similarly, treatment of non–small cell lung cancer cell lines with the EGFR inhibitors gefitinib and erlotinib induced increased DNA damage and temporary deficiency in base-excision repair (BER). The resulting increase in genetic instability promoted the acquisition of the EGFR p.T790M mutation, which drives acquired resistance to anti-EGFR treatment (76). In addition, DNA barcoding and mathematical modeling of triple-negative breast cancer cells in response to BET and CDK4/6 inhibitors showed a higher rate of de novo mutagenesis under drug pressure (77).

This growing body of literature suggests that the features of the stress response displayed by microbial organisms have counterparts in cancer cells, and supports the possibility that tumors are a community of individual entities with remarkable survival and evolutionary capabilities (5).

In E. coli, both RpoS-mediated general stress response and SOS DNA damage response need to be activated to initiate stress-induced adaptive mutability (45, 66). The SOS-mediated activation of error-prone DNA polymerases makes the repair of DSBs a source of mutagenesis and increased genetic variations (48). However, RpoS is required to permit the use of error-prone polymerases during stress (47). In addition, although the mechanisms of MMR downregulation during adaptive mutability are not well characterized, RpoS also seems to be implicated in this temporary impairment of DNA repair (78, 79). Importantly, RpoS, and not error-prone polymerases, appears to contribute to genetic amplification, suggesting a central role for RpoS in the induction of genome instability under stress (79).

Recently, MTOR has been proposed as the master regulator umpiring adaptive mutability in response to stress across multiple cancer types (5, 39, 64). Using genome-wide functional screens in response to different types of targeted therapies, Cipponi and colleagues identified two classes of genes involved in the development of secondary resistance to targeted therapy: (i) the “solution” genes, directly responsible for conferring resistance to the specific anticancer agent used (e.g., TP53 alterations in response to the MDM2 inhibitor nutlin); and (ii) “facilitator genes,” capable of indirectly facilitating the process of adaptation to drug-imposed hostile environment. MTOR was identified among the top-ranked facilitator genes, and its pharmacologic inhibition was shown to selectively impair HR and canonical high-fidelity DNA polymerase genes (39).

Moreover, Temprine and colleagues recently showed that treatment of melanoma cell lines with the BRAF inhibitor dabrafenib induced upregulation of error-prone polymerase κ (Pol κ), and this was central to the emergence of drug tolerance (64); furthermore, inhibition of mTOR recapitulated the phenotype (64). Polymerase κ is the mammalian ortholog of E. coli Pol IV, the error-prone DNA polymerase responsible for base substitutions and indels during mutagenic DNA break repair in E. coli (48). Altogether, these results suggest a key role for MTOR in fostering genetic diversity and promoting adaptive evolution in cancer cells during drug treatment.

Overcoming drug resistance remains a major challenge in oncology. Although it is generally accepted that the intrinsic molecular heterogeneity that characterizes most tumors plays a major role in the emergence of drug resistance, it remains unclear whether yet-to-be-discovered biological features of cancer cells play a role in therapeutic failures.

The analogies between drug-resistance mechanisms arising in cancer and infectious diseases, such as mutations in the drug target or activation/amplification of parallel compensatory pathways, suggest that they can provide insights for the development of novel anticancer treatments (80). For instance, the successful experience of combinatorial strategy at treatment initiation in the case of HIV and tuberculosis infections highlights the relevance of using drug combinations in order to achieve stronger tumor shrinkage, while preventing the outgrowth of resistant clones (80).

Here, we propose that further insights on how tumor cells survive adverse environments, and specifically stress generated by therapeutic treatment, can be obtained by observing the evolutionary strategies used by unicellular microorganisms in response to drug treatments and stressful environments (81), and could be translated into novel concepts for therapeutic interventions.

In detail, if stress response strategies exploited by unicellular organisms represent phylogenetically conserved mechanisms of survival, then conceptual interpretation of cancer as a community of unicellular organisms can be inspirational for developing new anticancer therapies to curb or restrict the emergence of drug resistance (81, 82).

Modern therapeutic regimens have often focused on targeting cell hyperproliferation features (e.g., targeting aberrant and constitutive activation of oncogenes). Though initially effective, activation of parallel pathways and/or compensatory feedback loops usually leads to treatment failure (83, 84), highlighting that cellular proliferation is a redundant, multilayer (and therefore seemingly unbeatable) process in cancer cells. However, by considering at least some features of cancer cells as a phylogenetic throwback, new vulnerabilities emerge. As a matter of fact, drugs targeting cellular processes shared with unicellular organisms are already used in clinical practice, including inhibitors of purine and pyrimidine synthesis, proteasome inhibitors, and inhibitors of the mitotic spindle (7).

Notably, like bacteria in response to antibiotics, cancer cells in quiescent/slowly replicating state may contribute to survival under stress, and represent a reservoir from which mechanisms of drug resistance could eventually emerge (54). Although both tumor progression and response to anticancer drugs are fueled by mutations, the evidence that patients can relapse after a long disease stabilization, or that tumors can be successfully rechallenged with the same drug, cannot be explained by genetic resistance. Dormant drug-tolerant tumor cells might represent a major source of relapse in these instances (53). The importance of targeting persister cells in both cancer and infectious disease in order to prevent relapse has been previously highlighted (80, 85, 86); however, little is known about persistence mechanisms.

We and others recently unveiled that cancer persister cells activate a stress-induced adaptive mutability response as an ancestral survival program that leads to increased mutagenicity, thus fueling selection of newly generated resistant variants (38, 39). Though adaptive mutability increases the chances of survival in stressful environments, this strategy comes at a cost, and stress-induced modulation of DNA-repair pathways might unveil cell vulnerabilities exploitable for innovative therapeutic strategies (81).

We propose here some therapeutic approaches aiming at preventing the development of drug resistance, targeting cancer cell features resembling the unicellular ones.

As discussed above, in bacteria and cancer cells undergoing adaptive mutability in response to stress, the MMR and HR DNA-repair machineries are repressed, and DNA damage is tolerated, thus promoting mutagenesis and improving survival. Interfering with these mechanisms may undermine cellular survival and promote DNA damage–induced cytotoxicity. Indeed, increasing levels of DNA damage to a point that cancer cells cannot tolerate is already exploited in clinical care. Tumors under therapeutic stress display increased endogenous DNA damage, together with delayed repair of radiation-induced and chemically induced DNA lesions, as pointed out by several works in the literature (Fig. 2; refs. 87–94). Indeed, targeted therapies are already successfully combined with radiotherapy or chemotherapy to increase chances of tumor control.

Figure 2.

Effect of targeted therapy–induced stress on the DNA-repair machinery in cancer cells. Treatment of cancer cells with several targeted therapies (indicated on the left with the corresponding molecular targets) results in enhanced DNA damage, accompanied by a promotion of tolerance over DNA damage recognition and repair (DDR), and upregulation of error-prone DNA polymerases. The concomitant downmodulation of the DNA-repair systems (depicted on the right) results in transient functional DDR deficiency which, in turn, is permissive for increased mutagenesis. While fostering genetic diversity and, therefore, survival, modulation of the DDR unveils possible vulnerabilities that might be exploited to eradicate cancer cells that undergo stress-induced adaptive evolution and foster the onset of resistance to targeted agents. The numbers in brackets indicate the references corresponding to each targeted treatment and associated DNA-repair modulation. NER, nucleotide excision repair.

Figure 2.

Effect of targeted therapy–induced stress on the DNA-repair machinery in cancer cells. Treatment of cancer cells with several targeted therapies (indicated on the left with the corresponding molecular targets) results in enhanced DNA damage, accompanied by a promotion of tolerance over DNA damage recognition and repair (DDR), and upregulation of error-prone DNA polymerases. The concomitant downmodulation of the DNA-repair systems (depicted on the right) results in transient functional DDR deficiency which, in turn, is permissive for increased mutagenesis. While fostering genetic diversity and, therefore, survival, modulation of the DDR unveils possible vulnerabilities that might be exploited to eradicate cancer cells that undergo stress-induced adaptive evolution and foster the onset of resistance to targeted agents. The numbers in brackets indicate the references corresponding to each targeted treatment and associated DNA-repair modulation. NER, nucleotide excision repair.

Close modal

Multiple pieces of evidence indicate modulation in DNA-repair systems upon treatment with targeted therapies across different tissues (Fig. 2; refs. 38, 39, 64, 76, 89, 91, 93–102). For example, blockade of the RAF/MEK/ERK pathway induces a BRCAness phenotype that sensitizes cells to PARP inhibitors (PARPi), both in vitro and in vivo (Fig. 2). Sun and colleagues showed that targeted therapy inhibiting the MAPK pathway restores sensitivity to PARPi in otherwise PARPi-resistant ovarian cancer cell lines (102). Relatedly, Maertens and colleagues found that cotreatment of melanoma cells with vemurafenib and histone deacetylases (HDAC) inhibitors leads to concomitant downregulation of both HR and NHEJ, an ineffective repair of DSBs, and ultimately cell catastrophe (ref. 101; Fig. 2). Based on this, synthetic lethality approaches capitalizing on the transient deficiency of DNA-repair systems in stressed cancer cells might be exploited to prevent cancer adaptation and evolution.

Analogously, inhibition of the transcriptional networks controlling induction of adaptive mutability in cancer cells might be successful in preventing development of resistance through de novo mutagenesis. Although MTOR has emerged as a possible master regulator of adaptive stress response in multiple tumors, it has been shown that mTOR inhibition recapitulates the adaptive mutability phenotype rather than preventing it (39). Therefore, the applicability of clinically available mTOR inhibitors for this purpose remains uncertain.

On the contrary, inhibition of hyperactivated error-prone DNA polymerases might eventually interfere with enhanced mutagenic capacity. For example, overexpression of Pol κ, which plays a central role in the adaptive response by regulating genes associated with drug resistance and immune surveillance through noncanonical mechanisms, further increases resistance to treatment, suggesting it as a putative target to curb drug tolerance (64).

Following the lesson learned from infectious disease, in order to optimize the potential success of these strategies in preventing cancer adaptation and curbing the tumor evolution, targeted therapies and drugs targeting the stress-response phenotype should be administered in combination from the initiation of the treatment (i.e., before development of resistance).

We and others previously reported the preclinical and clinical potential of concomitant inhibition of multiple nodes along the same pathway in terms of prevention of secondary resistance (83, 103) and overall survival in patients with colorectal cancer (104, 105).

Notably, as we previously demonstrated, interfering with oncogenic addiction causes increased DNA damage and shifts the dependency of cancer cells toward DDT pathways to cope with increased instability (38). The concomitant inhibition of multiple pathways with agents simultaneously targeting oncogene addiction and the adaptive response activated in cancer cells exposed to targeted therapies might therefore restrain the emergence of resistant variants by denying activation of the salvation mechanisms in cancer cells.

The concept of a paradigmatic shift from a reactive to a proactive strategy is indeed central to the approach of targeting mechanisms of adaptive mutability and resistance. Currently, in most instances medical oncologists wait for resistance to emerge; next, the resistant tumor is molecularly profiled, and then therapy is modified accordingly. Instead, one can envision acting preemptively by interfering with the cellular factors involved in adaptive mutability to restrain the emergence of resistance, rather than passively waiting for the tumor to evolve under therapy.

Although it remains unclear whether the stress-induced adaptive response also occurs in normal cells, inhibition of DNA damage response has been previously tested in patients with cancer with manageable toxicity (106). Moreover, combinatorial regimens might allow the administration of lower doses of single agents by exploiting synergism exerted by parallel inhibition of multiple survival pathways in cancer cells.

In conclusion, further exploration of the mechanisms through which bacteria and cancer cells survive and evolve under stress could provide new therapeutic options aimed at curbing adaptive mutability unleashed by drug pressures in cancer cells with altered genome functions, while limiting the side effects on normal cells.

A. Bardelli reports grants from AIRC during the conduct of the study; grants from Neophore and AstraZeneca, and personal fees from Inivata outside the submitted work; SAB at Horizon Discovery, Inivata, and Roche; and shareholding at Neophore. No disclosures were reported by the other authors.

This work was supported by FONDAZIONE AIRC under 5 per Mille 2018—ID. 21091 program—P.I. Alberto Bardelli; AIRC under IG 2018—ID. 21923 project—P.I. Alberto Bardelli; AIRC-CRUK-FC AECC Accelerator Award contract 22795 to A. Bardelli; IMI contract n. 101007937 PERSIST-SEQ to A. Bardelli; Ministero Salute, RC 2020 to A. Bardelli.

1.
Aktipis
CA
,
Boddy
AM
,
Jansen
G
,
Hibner
U
,
Hochberg
ME
,
Maley
CC
, et al
Cancer across the tree of life: cooperation and cheating in multicellularity
.
Philos Trans R Soc Lond B Biol Sci
2015
;
370
:
20140219
.
2.
Hanahan
D
,
Weinberg
RA
. 
Hallmarks of cancer: the next generation
.
Cell
2011
;
144
:
646
74
.
3.
Lambert
G
,
Estévez-Salmeron
L
,
Oh
S
,
Liao
D
,
Emerson
BM
,
Tlsty
TD
, et al
An analogy between the evolution of drug resistance in bacterial communities and malignant tissues
.
Nat Rev Cancer
2011
;
11
:
375
82
.
4.
Ben-Jacob
E
,
Coffey
DS
,
Levine
H
. 
Bacterial survival strategies suggest rethinking cancer cooperativity
.
Trends Microbiol
2012
;
20
:
403
10
.
5.
Fitzgerald
DM
,
Hastings
PJ
,
Rosenberg
SM
. 
Stress-induced mutagenesis: implications in cancer and drug resistance
.
Annu Rev Cancer Biol
2017
;
1
:
119
40
.
6.
Rosenberg
SM
,
Shee
C
,
Frisch
RL
,
Hastings
PJ
. 
Stress-induced mutation via DNA breaks in Escherichia coli: a molecular mechanism with implications for evolution and medicine
.
Bioessays
2012
;
34
:
885
92
.
7.
Trigos
AS
,
Pearson
RB
,
Papenfuss
AT
,
Goode
DL
. 
How the evolution of multicellularity set the stage for cancer
.
Br J Cancer
2018
;
118
:
145
52
.
8.
Vincent
M
. 
Cancer: a de-repression of a default survival program common to all cells?: a life-history perspective on the nature of cancer
.
Bioessays
2012
;
34
:
72
82
.
9.
Lineweaver
CH
,
Davies
PC
,
Vincent
MD
. 
Targeting cancer's weaknesses (not its strengths): therapeutic strategies suggested by the atavistic model
.
Bioessays
2014
;
36
:
827
35
.
10.
Hibbing
ME
,
Fuqua
C
,
Parsek
MR
,
Peterson
SB
. 
Bacterial competition: surviving and thriving in the microbial jungle
.
Nat Rev Microbiol
2010
;
8
:
15
25
.
11.
Nadell
CD
,
Drescher
K
,
Foster
KR
. 
Spatial structure, cooperation and competition in biofilms
.
Nat Rev Microbiol
2016
;
14
:
589
600
.
12.
Sun
R
,
Hu
Z
,
Sottoriva
A
,
Graham
TA
,
Harpak
A
,
Ma
Z
, et al
Between-region genetic divergence reflects the mode and tempo of tumor evolution
.
Nat Genet
2017
;
49
:
1015
24
.
13.
Siravegna
G
,
Mussolin
B
,
Buscarino
M
,
Corti
G
,
Cassingena
A
,
Crisafulli
G
, et al
Clonal evolution and resistance to EGFR blockade in the blood of colorectal cancer patients
.
Nat Med
2015
;
21
:
795
801
.
14.
McGranahan
N
,
Swanton
C
. 
Clonal heterogeneity and tumor evolution: past, present, and the future
.
Cell
2017
;
168
:
613
28
.
15.
Claessen
D
,
Rozen
DE
,
Kuipers
OP
,
Søgaard-Andersen
L
,
van Wezel
GP
. 
Bacterial solutions to multicellularity: a tale of biofilms, filaments and fruiting bodies
.
Nat Rev Microbiol
2014
;
12
:
115
24
.
16.
Monds
RD
,
O'Toole
GA
. 
The developmental model of microbial biofilms: ten years of a paradigm up for review
.
Trends Microbiol
2009
;
17
:
73
87
.
17.
Sahai
E
,
Astsaturov
I
,
Cukierman
E
,
DeNardo
DG
,
Egeblad
M
,
Evans
RM
, et al
A framework for advancing our understanding of cancer-associated fibroblasts
.
Nat Rev Cancer
2020
;
20
:
174
86
.
18.
Binnewies
M
,
Roberts
EW
,
Kersten
K
,
Chan
V
,
Fearon
DF
,
Merad
M
, et al
Understanding the tumor immune microenvironment (TIME) for effective therapy
.
Nat Med
2018
;
24
:
541
50
.
19.
Hobor
S
,
Van Emburgh
BO
,
Crowley
E
,
Misale
S
,
Di Nicolantonio
F
,
Bardelli
A
. 
TGFα and amphiregulin paracrine network promotes resistance to EGFR blockade in colorectal cancer cells
.
Clin Cancer Res
2014
;
20
:
6429
38
.
20.
Butler
MT
,
Wang
Q
,
Harshey
RM
. 
Cell density and mobility protect swarming bacteria against antibiotics
.
Proc Natl Acad Sci U S A
2010
;
107
:
3776
81
.
21.
Branda
SS
,
González-Pastor
JE
,
Ben-Yehuda
S
,
Losick
R
,
Kolter
R
. 
Fruiting body formation by Bacillus subtilis
.
Proc Natl Acad Sci U S A
2001
;
98
:
11621
6
.
22.
Valastyan
S
,
Weinberg
RA
. 
Tumor metastasis: molecular insights and evolving paradigms
.
Cell
2011
;
147
:
275
92
.
23.
Hüsemann
Y
,
Geigl
JB
,
Schubert
F
,
Musiani
P
,
Meyer
M
,
Burghart
E
, et al
Systemic spread is an early step in breast cancer
.
Cancer Cell
2008
;
13
:
58
68
.
24.
Sprouffske
K
,
Merlo
LM
,
Gerrish
PJ
,
Maley
CC
,
Sniegowski
PD
. 
Cancer in light of experimental evolution
.
Curr Biol
2012
;
22
:
R762
71
.
25.
Matic
I
,
Radman
M
,
Taddei
F
,
Picard
B
,
Doit
C
,
Bingen
E
, et al
Highly variable mutation rates in commensal and pathogenic Escherichia coli
.
Science
1997
;
277
:
1833
4
.
26.
Mao
EF
,
Lane
L
,
Lee
J
,
Miller
JH
. 
Proliferation of mutators in a cell population
.
J Bacteriol
1997
;
179
:
417
22
.
27.
Taddei
F
,
Radman
M
,
Maynard-Smith
J
,
Toupance
B
,
Gouyon
PH
,
Godelle
B
. 
Role of mutator alleles in adaptive evolution
.
Nature
1997
;
387
:
700
2
.
28.
Tenaillon
O
,
Taddei
F
,
Radmian
M
,
Matic
I
. 
Second-order selection in bacterial evolution: selection acting on mutation and recombination rates in the course of adaptation
.
Res Microbiol
2001
;
152
:
11
6
.
29.
Tanaka
MM
,
Bergstrom
CT
,
Levin
BR
. 
The evolution of mutator genes in bacterial populations: the roles of environmental change and timing
.
Genetics
2003
;
164
:
843
54
.
30.
Davies
PC
,
Lineweaver
CH
. 
Cancer tumors as Metazoa 1.0: tapping genes of ancient ancestors
.
Phys Biol
2011
;
8
:
015001
.
31.
Trigos
AS
,
Pearson
RB
,
Papenfuss
AT
,
Goode
DL
. 
Altered interactions between unicellular and multicellular genes drive hallmarks of transformation in a diverse range of solid tumors
.
Proc Natl Acad Sci U S A
2017
;
114
:
6406
11
.
32.
Wu
A
,
Zhang
Q
,
Lambert
G
,
Khin
Z
,
Gatenby
RA
,
Kim
HJ
, et al
Ancient hot and cold genes and chemotherapy resistance emergence
.
Proc Natl Acad Sci U S A
2015
;
112
:
10467
72
.
33.
Chen
H
,
He
X
. 
The convergent cancer evolution toward a single cellular destination
.
Mol Biol Evol
2016
;
33
:
4
12
.
34.
Chen
H
,
Lin
F
,
Xing
K
,
He
X
. 
The reverse evolution from multicellularity to unicellularity during carcinogenesis
.
Nat Commun
2015
;
6
:
6367
.
35.
Cisneros
L
,
Bussey
KJ
,
Orr
AJ
,
Miočević
M
,
Lineweaver
CH
,
Davies
P
. 
Ancient genes establish stress-induced mutation as a hallmark of cancer
.
PLoS One
2017
;
12
:
e0176258
.
36.
Trigos
AS
,
Pearson
RB
,
Papenfuss
AT
,
Goode
DL
. 
Somatic mutations in early metazoan genes disrupt regulatory links between unicellular and multicellular genes in cancer
.
Elife
2019
;
8
:
e40947
.
37.
Thomas
F
,
Ujvari
B
,
Renaud
F
,
Vincent
M
. 
Cancer adaptations: atavism, de novo selection, or something in between?
Bioessays
2017
;
39
:
8
.
38.
Russo
M
,
Crisafulli
G
,
Sogari
A
,
Reilly
NM
,
Arena
S
,
Lamba
S
, et al
Adaptive mutability of colorectal cancers in response to targeted therapies
.
Science
2019
;
366
:
1473
80
.
39.
Cipponi
A
,
Goode
DL
,
Bedo
J
,
McCabe
MJ
,
Pajic
M
,
Croucher
DR
, et al
MTOR signaling orchestrates stress-induced mutagenesis, facilitating adaptive evolution in cancer
.
Science
2020
;
368
:
1127
31
.
40.
Rosenberg
SM
. 
Evolving responsively: adaptive mutation
.
Nat Rev Genet
2001
;
2
:
504
15
.
41.
Foster
PL
. 
Adaptive mutation: implications for evolution
.
Bioessays
2000
;
22
:
1067
74
.
42.
Balaban
NQ
,
Merrin
J
,
Chait
R
,
Kowalik
L
,
Leibler
S
. 
Bacterial persistence as a phenotypic switch
.
Science
2004
;
305
:
1622
5
.
43.
Harms
A
,
Maisonneuve
E
,
Gerdes
K
. 
Mechanisms of bacterial persistence during stress and antibiotic exposure
.
Science
2016
;
354
:
aaf4268
.
44.
Gottesman
S
. 
Trouble is coming: signaling pathways that regulate general stress responses in bacteria
.
J Biol Chem
2019
;
294
:
11685
700
.
45.
Foster
PL
. 
Stress-induced mutagenesis in bacteria
.
Crit Rev Biochem Mol Biol
2007
;
42
:
373
97
.
46.
Sutton
MD
,
Smith
BT
,
Godoy
VG
,
Walker
GC
. 
The SOS response: recent insights into umuDC-dependent mutagenesis and DNA damage tolerance
.
Annu Rev Genet
2000
;
34
:
479
97
.
47.
Ponder
RG
,
Fonville
NC
,
Rosenberg
SM
. 
A switch from high-fidelity to error-prone DNA double-strand break repair underlies stress-induced mutation
.
Mol Cell
2005
;
19
:
791
804
.
48.
McKenzie
GJ
,
Lee
PL
,
Lombardo
MJ
,
Hastings
PJ
,
Rosenberg
SM
. 
SOS mutator DNA polymerase IV functions in adaptive mutation and not adaptive amplification
.
Mol Cell
2001
;
7
:
571
9
.
49.
Goodman
MF
. 
Error-prone repair DNA polymerases in prokaryotes and eukaryotes
.
Annu Rev Biochem
2002
;
71
:
17
50
.
50.
Battesti
A
,
Majdalani
N
,
Gottesman
S
. 
The RpoS-mediated general stress response in Escherichia coli
.
Annu Rev Microbiol
2011
;
65
:
189
213
.
51.
Sharma
SV
,
Lee
DY
,
Li
B
,
Quinlan
MP
,
Takahashi
F
,
Maheswaran
S
, et al
A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations
.
Cell
2010
;
141
:
69
80
.
52.
Hata
AN
,
Niederst
MJ
,
Archibald
HL
,
Gomez-Caraballo
M
,
Siddiqui
FM
,
Mulvey
HE
, et al
Tumor cells can follow distinct evolutionary paths to become resistant to epidermal growth factor receptor inhibition
.
Nat Med
2016
;
22
:
262
9
.
53.
Shen
S
,
Vagner
S
,
Robert
C
. 
Persistent cancer cells: the deadly survivors
.
Cell
2020
;
183
:
860
74
.
54.
Ramirez
M
,
Rajaram
S
,
Steininger
RJ
,
Osipchuk
D
,
Roth
MA
,
Morinishi
LS
, et al
Diverse drug-resistance mechanisms can emerge from drug-tolerant cancer persister cells
.
Nat Commun
2016
;
7
:
10690
.
55.
Saxton
RA
,
Sabatini
DM
. 
mTOR signaling in growth, metabolism, and disease
.
Cell
2017
;
168
:
960
76
.
56.
Tissier
A
,
McDonald
JP
,
Frank
EG
,
Woodgate
R
. 
poliota, a remarkably error-prone human DNA polymerase
.
Genes Dev
2000
;
14
:
1642
50
.
57.
Rattray
AJ
,
Strathern
JN
. 
Error-prone DNA polymerases: when making a mistake is the only way to get ahead
.
Annu Rev Genet
2003
;
37
:
31
66
.
58.
Ghosal
G
,
Chen
J
. 
DNA damage tolerance: a double-edged sword guarding the genome
.
Transl Cancer Res
2013
;
2
:
107
29
.
59.
Seki
M
,
Gearhart
PJ
,
Wood
RD
. 
DNA polymerases and somatic hypermutation of immunoglobulin genes
.
EMBO Rep
2005
;
6
:
1143
8
.
60.
Yoon
JH
,
McArthur
MJ
,
Park
J
,
Basu
D
,
Wakamiya
M
,
Prakash
L
, et al
Error-prone replication through UV lesions by DNA polymerase θ protects against skin cancers
.
Cell
2019
;
176
:
1295
309
.
61.
Breivik
J
,
Gaudernack
G
. 
Resolving the evolutionary paradox of genetic instability: a cost-benefit analysis of DNA repair in changing environments
.
FEBS Lett
2004
;
563
:
7
12
.
62.
O-Wang
J
,
Kawamura
K
,
Tada
Y
,
Ohmori
H
,
Kimura
H
,
Sakiyama
S
, et al
DNA polymerase kappa, implicated in spontaneous and DNA damage-induced mutagenesis, is overexpressed in lung cancer
.
Cancer Res
2001
;
61
:
5366
9
.
63.
Wang
H
,
Wu
W
,
Wang
HW
,
Wang
S
,
Chen
Y
,
Zhang
X
, et al
Analysis of specialized DNA polymerases expression in human gliomas: association with prognostic significance
.
Neuro Oncol
2010
;
12
:
679
86
.
64.
Temprine
K
,
Campbell
NR
,
Huang
R
,
Langdon
EM
,
Simon-Vermot
T
,
Mehta
K
, et al
Regulation of the error-prone DNA polymerase Polκ by oncogenic signaling and its contribution to drug resistance
.
Sci Signal
2020
;
13
:
eaau1453
.
65.
Lipinski
KA
,
Barber
LJ
,
Davies
MN
,
Ashenden
M
,
Sottoriva
A
,
Gerlinger
M
. 
Cancer evolution and the limits of predictability in precision cancer medicine
.
Trends Cancer
2016
;
2
:
49
63
.
66.
Galhardo
RS
,
Hastings
PJ
,
Rosenberg
SM
. 
Mutation as a stress response and the regulation of evolvability
.
Crit Rev Biochem Mol Biol
2007
;
42
:
399
435
.
67.
Gonzalez
C
,
Hadany
L
,
Ponder
RG
,
Price
M
,
Hastings
PJ
,
Rosenberg
SM
. 
Mutability and importance of a hypermutable cell subpopulation that produces stress-induced mutants in Escherichia coli
.
PLoS Genet
2008
;
4
:
e1000208
.
68.
Shee
C
,
Gibson
JL
,
Rosenberg
SM
. 
Two mechanisms produce mutation hotspots at DNA breaks in Escherichia coli
.
Cell Rep
2012
;
2
:
714
21
.
69.
Roberts
SA
,
Gordenin
DA
. 
Hypermutation in human cancer genomes: footprints and mechanisms
.
Nat Rev Cancer
2014
;
14
:
786
800
.
70.
Gutierrez
A
,
Laureti
L
,
Crussard
S
,
Abida
H
,
Rodríguez-Rojas
A
,
Blázquez
J
, et al
β-Lactam antibiotics promote bacterial mutagenesis via an RpoS-mediated reduction in replication fidelity
.
Nat Commun
2013
;
4
:
1610
.
71.
Kohanski
MA
,
DePristo
MA
,
Collins
JJ
. 
Sublethal antibiotic treatment leads to multidrug resistance via radical-induced mutagenesis
.
Mol Cell
2010
;
37
:
311
20
.
72.
Pal
D
,
Pertot
A
,
Shirole
NH
,
Yao
Z
,
Anaparthy
N
,
Garvin
T
, et al
TGF-β reduces DNA ds-break repair mechanisms to heighten genetic diversity and adaptability of CD44+/CD24- cancer cells
.
Elife
2017
;
6
:
e21615
.
73.
Scanlon
SE
,
Glazer
PM
. 
Multifaceted control of DNA repair pathways by the hypoxic tumor microenvironment
.
DNA Repair
2015
;
32
:
180
9
.
74.
Mihaylova
VT
,
Bindra
RS
,
Yuan
J
,
Campisi
D
,
Narayanan
L
,
Jensen
R
, et al
Decreased expression of the DNA mismatch repair gene Mlh1 under hypoxic stress in mammalian cells
.
Mol Cell Biol
2003
;
23
:
3265
73
.
75.
Tomasetti
C
,
Li
L
,
Vogelstein
B
. 
Stem cell divisions, somatic mutations, cancer etiology, and cancer prevention
.
Science
2017
;
355
:
1330
4
.
76.
Cao
X
,
Zhou
Y
,
Sun
H
,
Xu
M
,
Bi
X
,
Zhao
Z
, et al
EGFR-TKI-induced HSP70 degradation and BER suppression facilitate the occurrence of the EGFR T790 M resistant mutation in lung cancer cells
.
Cancer Lett
2018
;
424
:
84
96
.
77.
Ge
JY
,
Shu
S
,
Kwon
M
,
Jovanović
B
,
Murphy
K
,
Gulvady
A
, et al
Acquired resistance to combined BET and CDK4/6 inhibition in triple-negative breast cancer
.
Nat Commun
2020
;
11
:
2350
.
78.
McKenzie
GJ
,
Harris
RS
,
Lee
PL
,
Rosenberg
SM
. 
The SOS response regulates adaptive mutation
.
Proc Natl Acad Sci U S A
2000
;
97
:
6646
51
.
79.
McKenzie
GJ
,
Rosenberg
SM
. 
Adaptive mutations, mutator DNA polymerases and genetic change strategies of pathogens
.
Curr Opin Microbiol
2001
;
4
:
586
94
.
80.
Glickman
MS
,
Sawyers
CL
. 
Converting cancer therapies into cures: lessons from infectious diseases
.
Cell
2012
;
148
:
1089
98
.
81.
Rosenberg
SM
,
Queitsch
C
. 
Medicine. Combating evolution to fight disease
.
Science
2014
;
343
:
1088
9
.
82.
Al Mamun
AA
,
Lombardo
MJ
,
Shee
C
,
Lisewski
AM
,
Gonzalez
C
,
Lin
D
, et al
Identity and function of a large gene network underlying mutagenic repair of DNA breaks
.
Science
2012
;
338
:
1344
8
.
83.
Prahallad
A
,
Sun
C
,
Huang
S
,
Di Nicolantonio
F
,
Salazar
R
,
Zecchin
D
, et al
Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR
.
Nature
2012
;
483
:
100
3
.
84.
Amodio
V
,
Yaeger
R
,
Arcella
P
,
Cancelliere
C
,
Lamba
S
,
Lorenzato
A
, et al
EGFR blockade reverts resistance to KRAS
.
Cancer Discov
2020
;
10
:
1129
39
.
85.
Hangauer
MJ
,
Viswanathan
VS
,
Ryan
MJ
,
Bole
D
,
Eaton
JK
,
Matov
A
, et al
Drug-tolerant persister cancer cells are vulnerable to GPX4 inhibition
.
Nature
2017
;
551
:
247
50
.
86.
De Conti
G
,
Dias
MH
,
Bernards
R
. 
Fighting drug resistance through the targeting of drug-tolerant persister cells
.
Cancers
2021
;
13
:
1118
.
87.
Chinnaiyan
P
,
Huang
S
,
Vallabhaneni
G
,
Armstrong
E
,
Varambally
S
,
Tomlins
SA
, et al
Mechanisms of enhanced radiation response following epidermal growth factor receptor signaling inhibition by erlotinib (Tarceva)
.
Cancer Res
2005
;
65
:
3328
35
.
88.
Tanaka
T
,
Munshi
A
,
Brooks
C
,
Liu
J
,
Hobbs
ML
,
Meyn
RE
. 
Gefitinib radiosensitizes non–small cell lung cancer cells by suppressing cellular DNA repair capacity
.
Clin Cancer Res
2008
;
14
:
1266
.
89.
Dittmann
K
,
Mayer
C
,
Fehrenbacher
B
,
Schaller
M
,
Raju
U
,
Milas
L
, et al
Radiation-induced epidermal growth factor receptor nuclear import is linked to activation of DNA-dependent protein kinase
.
J Biol Chem
2005
;
280
:
31182
9
.
90.
Lim
J
,
Yang
K
,
Taylor-Harding
B
,
Wiedemeyer
WR
,
Buckanovich
RJ
. 
VEGFR3 inhibition chemosensitizes ovarian cancer stemlike cells through down-regulation of BRCA1 and BRCA2
.
Neoplasia
2014
;
16
:
343
53
.
91.
Robb
R
,
Yang
L
,
Shen
C
,
Wolfe
AR
,
Webb
A
,
Zhang
X
, et al
Inhibiting BRAF oncogene-mediated radioresistance effectively radiosensitizes BRAF
.
Clin Cancer Res
2019
;
25
:
4749
60
.
92.
Liu
J
,
Jiang
G
,
Mao
P
,
Zhang
J
,
Zhang
L
,
Liu
L
, et al
Down-regulation of GADD45A enhances chemosensitivity in melanoma
.
Sci Rep
2018
;
8
:
4111
.
93.
Li
W
,
Melton
DW
. 
Cisplatin regulates the MAPK kinase pathway to induce increased expression of DNA repair gene ERCC1 and increase melanoma chemoresistance
.
Oncogene
2012
;
31
:
2412
22
.
94.
Estrada-Bernal
A
,
Chatterjee
M
,
Haque
SJ
,
Yang
L
,
Morgan
MA
,
Kotian
S
, et al
MEK inhibitor GSK1120212-mediated radiosensitization of pancreatic cancer cells involves inhibition of DNA double-strand break repair pathways
.
Cell Cycle
2015
;
14
:
3713
24
.
95.
Nowsheen
S
,
Bonner
JA
,
LoBuglio
AF
,
Trummell
H
,
Whitley
AC
,
Dobelbower
MC
, et al
Cetuximab augments cytotoxicity with poly (ADP-Ribose) polymerase inhibition in head and neck cancer
.
PLoS One
2011
;
6
:
e24148
.
96.
Li
L
,
Wang
H
,
Yang
ES
,
Arteaga
CL
,
Xia
F
. 
Erlotinib attenuates homologous recombinational repair of chromosomal breaks in human breast cancer cells
.
Cancer Res
2008
;
68
:
9141
.
97.
Friedmann
BJ
,
Caplin
M
,
Savic
B
,
Shah
T
,
Lord
CJ
,
Ashworth
A
, et al
Interaction of the epidermal growth factor receptor and the DNA-dependent protein kinase pathway following gefitinib treatment
.
Mol Cancer Ther
2006
;
5
:
209
.
98.
Ibrahim
YH
,
García-García
C
,
Serra
V
,
He
L
,
Torres-Lockhart
K
,
Prat
A
, et al
PI3K inhibition impairs BRCA1/2 expression and sensitizes BRCA-proficient triple-negative breast cancer to PARP inhibition
.
Cancer Discov
2012
;
2
:
1036
.
99.
Juvekar
A
,
Burga
LN
,
Hu
H
,
Lunsford
EP
,
Ibrahim
YH
,
Balmañà
J
, et al
Combining a PI3K inhibitor with a PARP inhibitor provides an effective therapy for BRCA1-related breast cancer
.
Cancer Discov
2012
;
2
:
1048
.
100.
Mo
W
,
Liu
Q
,
Lin
CC-J
,
Dai
H
,
Peng
Y
,
Liang
Y
, et al
mTOR inhibitors suppress homologous recombination repair and synergize with PARP inhibitors via regulating SUV39H1 in BRCA-proficient triple-negative breast cancer
.
Clin Cancer Res
2016
;
22
:
1699
.
101.
Maertens
O
,
Kuzmickas
R
,
Manchester
HE
,
Emerson
CE
,
Gavin
AG
,
Guild
CJ
, et al
MAPK pathway suppression unmasks latent DNA repair defects and confers a chemical synthetic vulnerability in BRAF-, NRAS-, and NF1-mutant melanomas
.
Cancer Discov
2019
;
9
:
526
.
102.
Sun
C
,
Fang
Y
,
Yin
J
,
Chen
J
,
Ju
Z
,
Zhang
D
, et al
Rational combination therapy with PARP and MEK inhibitors capitalizes on therapeutic liabilities in RAS mutant cancers
.
Sci Transl Med
2017
;
9
:
eaal5148
.
103.
Misale
S
,
Arena
S
,
Lamba
S
,
Siravegna
G
,
Lallo
A
,
Hobor
S
, et al
Blockade of EGFR and MEK intercepts heterogeneous mechanisms of acquired resistance to Anti-EGFR therapies in colorectal cancer
.
Sci Transl Med
2014
;
6
:
224ra26
.
104.
Sartore-Bianchi
A
,
Trusolino
L
,
Martino
C
,
Bencardino
K
,
Lonardi
S
,
Bergamo
F
, et al
Dual-targeted therapy with trastuzumab and lapatinib in treatment-refractory, KRAS codon 12/13 wild-type, HER2-positive metastatic colorectal cancer (HERACLES): a proof-of-concept, multicentre, open-label, phase 2 trial
.
Lancet Oncol
2016
;
17
:
738
46
.
105.
Kopetz
S
,
Grothey
A
,
Yaeger
R
,
Van Cutsem
E
,
Desai
J
,
Yoshino
T
, et al
Encorafenib, binimetinib, and cetuximab in
.
N Engl J Med
2019
;
381
:
1632
43
.
106.
Pilié
PG
,
Tang
C
,
Mills
GB
,
Yap
TA
. 
State-of-the-art strategies for targeting the DNA damage response in cancer
.
Nat Rev Clin Oncol
2019
;
16
:
81
104
.