One of the main reasons we have not been able to cure cancers is that treatments select for drug-resistant cells. Pest managers face similar challenges with pesticides selecting for pesticide-resistant insects, resulting in similar mechanisms of resistance. Pest managers have developed 10 principles that could be translated to controlling cancers: (i) prevent onset, (ii) monitor continuously, (iii) identify thresholds below which there will be no intervention, (iv) change interventions in response to burden, (v) preferentially select nonchemical control methods, (vi) use target-specific drugs, (vii) use the lowest effective dose, (viii) reduce cross-resistance, (ix) evaluate success based on long-term management, and (x) forecast growth and response. These principles are general to all cancers and cancer drugs and so could be employed broadly to improve oncology. Here, we review the parallel difficulties in controlling drug resistance in pests and cancer cells. We show how the principles of resistance management in pests might be applied to cancer. Integrated pest management inspired the development of adaptive therapy in oncology to increase progression-free survival and quality of life in patients with cancers where cures are unlikely. These pest management principles have the potential to inform clinical trial design.

Oncologists and pest managers have similar problems in that both cancer cells and pest populations are composed of large numbers of genetically diverse organisms spread over heterogeneous (micro)environments (13). When pest managers apply a pesticide to kill pests and oncologists apply a drug to kill cancer cells, they kill the sensitive pests and cancer cells but select for variants that are resistant to the drug (4, 5). In fact, most deaths from cancer are caused by the evolution of therapeutic resistance (6, 7). Both fields also face the challenge of limiting collateral damage and other negative side effects due to the toxicities of their drugs (8, 9). Pesticides have toxic effects on nonpest organisms in the environment and pose a threat to human consumers of the crops, analogous to the toxic effects of anticancer drugs on normal (noncancer) cells and the health of the patient (10, 11).

To date, almost all cancer therapies select for therapeutic resistance (7). Failure to address the evolutionary dynamics of cancer results in the sustained high mortality of metastatic disease and doom future cancer therapies to the same fate. Decades of experimentation in pest management have led to a series of insights and effective heuristics for controlling pests, called integrated pest management (IPM) that, for the most part, have never been tried in oncology. What can oncologists learn from pest managers and how might we translate those insights to the clinic?

IPM is a comprehensive approach to pest management emphasizing multifaceted, system-based strategies involving multiple disciplines (8, 1215). IPM was initially introduced as “integrated control” in 1959 by Stern and colleagues (16), who synthesized aspects of biological, spatial, and chemical control tactics to reduce pest populations below levels of economic concern while limiting the development of resistance within agricultural pest communities.

Translated to oncology, an analogous approach would be to control cancers so that patients can live with the disease, but not die from it. Although this might seem like a radical change in how we treat cancer, it is already the de facto goal in many of the treatments of metastatic cancers in which cures are extremely unlikely (17, 18). The decision of whether to treat with intent to cure or treat with intent to control should depend on evidence for the curability of the cancer as well as the age of the patient, their frailty, and the potential morbidities associated with the different kinds of treatment. Control would likely be more palatable in an elderly patient compared with a pediatric patient.

Here, we briefly review the history of the emergence of acquired therapeutic resistance and early attempts to address it by using multiple drugs in both pest management and oncology. We then review the principles of IPM (19) and discuss how they might be translated to oncology. Finally, we review early promising results in the application of those principles to oncology and provide an example of a potential future clinical trial in colorectal cancer based on IPM.

As early as 1914, pest managers observed that single-agent pesticide treatments did not work long-term because they selected for pesticide resistance (10). New insecticides developed in the 1960s and 70s were initially highly effective but quickly lost their effectiveness due to insects evolving mechanisms of resistance (2022). Insects even became resistant to alternative methods of control like crop rotation (23, 24).

Acquired therapeutic resistance was also documented in the first clinical reports of chemotherapy for cancer (25, 26). Single-drug therapies, such as nitrogen mustard and aminopterin, were used in the treatment of tumors in the late 1940s (25, 26). However, acquired resistance to treatment was observed for both drugs (25, 26). Resistance also evolved in response to radiotherapy (27). This helped drive interest in developing chemotherapies as the main method to treat cancer (25).

Targeted therapies, including both small molecules and biologics, also select for resistance (7, 28). Perhaps the most famous targeted therapy, imatinib mesylate, eventually leads to resistance in chronic myelogenous leukemia, gastrointestinal stromal tumors, and in Philadelphia chromosome–positive acute lymphoblastic leukemia (29). Trastuzumab, targeting HER2+ breast cancer, only worked for a median of 9 months in one study (30) and 4.9 months in another study, with the majority of patients developing resistance within a year (31). Targeted therapies for EGFR are notorious for selecting for resistance mutations. Most patients with non–small cell lung cancer (NSCLC) relapse on gefitinib (32), erlotinib (33), osimertinib (34), or crizotinib (35).

Recently immunotherapies, in particular immune checkpoint blockade therapies, have received well-deserved attention and excitement for generating responses, and sometimes apparent cures in some of the most difficult kinds of cancer to treat: lung cancers and melanoma. However, they also often select for acquired therapeutic resistance (3638). In melanoma, between a quarter to a half of patients acquire resistance and progress within 5 years on immune checkpoint blockade therapy (3640), and as many as 65% of patients with NSCLC progress within 4 years (41).

Multidrug therapies are often better than single drugs but still tend to fail, particularly in late-stage disease (4247). In theory, if resistance to a single drug is rare in a population of pests or cancer cells and the mechanism of resistance is different for each drug, resistance to multiple independent drugs should be much rarer than resistance to a single drug. Experience has shown that this is not the case. Even though several studies have shown that combined therapy is better than monotherapy in both pest management and cancer therapy, combined therapy still selects for multidrug resistance and ultimately fails (4248). This is in part due to cross-resistance between drugs and mechanisms of multidrug resistance, such as the upregulation of efflux pumps that can provide resistance to drugs with independent modes of action (MoA; refs. 45, 49, 50). Therefore, in most cases, combining drugs has not solved the problem of acquired therapeutic resistance.

Evidence in pest management

In 1989, Tabashnik provided a detailed review of the failures of multiple drug usage in pest management (5). The combinatorial use of insecticides in several studies showed that this method could not prevent resistance significantly (4244, 5153). There is an extensive and old literature in pest management that discusses different ways to calculate and detect synergy (and antagonism) between drugs, most of which depend on generating a model of the expected effect of the combination and comparing it to the observed effect (54). Plana and colleagues (55) have applied similar methods for defining (and finding very little) synergy in cancer drug combinations. There are important concerns about the combinatorial use of drugs, including toxicity for the natural predators of the pests, development of resistance in secondary pests, and increased selective pressure for cross-resistance (5).

Ozaki and colleagues (42) combined pesticides with potentially synergistic effects and different MoAs. They found that combinations of two or more insecticides either together, or alternating single drugs could delay the development of resistance, though multi-insecticide resistance still evolved (42). MacDonald and colleagues (44) tested the use of single insecticides to control house flies and also used multiple drugs in rotation. Overall, resistance most rapidly emerged in the single-drug treatment groups. Alternating drugs reduced the evolution of resistance and delayed resistance in the field (44). In practice, even theoretically synergistic combinations of pesticides are selected for multidrug resistance (14).

Overall, even though combining insecticides remains a theoretically justified tactic for delaying resistance in the case of pest management, mixtures that meet the optimization criteria of models remain elusive in practice (52).

Evidence in late-stage cancer therapy

Early-stage cancer is often curable through resection, and survival rates are typically much higher than late-stage cancers (56). The standard of care in many late-stage cancer treatments is to combine drugs (50). It is widely acknowledged that single-drug therapies tend to fail in cancer. What is less widely acknowledged is that multidrug therapies also tend to fail, particularly in late-stage adult cancers, due to multidrug resistance (45). Meta-analyses of lung (50, 5759), breast (60), gastric (60, 61), pancreatic (6264), and ovarian cancers (65, 66) show modest survival benefits of combining drugs with increased toxicity but rarely cures from complex combined therapies.

A meta-analysis of 35 randomized phase III trials in NSCLC compared single-drug therapy versus two or three drugs (50). There was a significant increase in both tumor response [OR, 0.42; 95% confidence interval (CI), 0.37–0.47; P < 0.001] and 1-year survival (OR, 0.80; 95% CI, 0.70–0.91; P < 0.001) using doublet regimes compared with single-drug therapy. In addition, they found an increase in the tumor response rate when three drugs were used in combination (OR, 0.66; 95% CI, 0.58–0.75; P < 0.001). However, there was no increase in 1-year survival when the third drug was added (OR, 1.01; 95% CI, 0.85–1.21; P = 0.88). Overall, greater toxicity was also observed in combination chemotherapy compared with single-agent chemotherapy (50). Grade 3 and 4 toxicity rates were higher with doublet regimens compared with single-agent therapy (ORs 1.2–6.2), as well as in triplet regimens compared with doublet regimens (ORs 1.4–2.9; ref. 50). A previous meta-analysis of single versus combination therapy for NSCLC found a 22% increase in 1-year survival with combination therapy compared with single-agent chemotherapy (RR: 1.22; 95% CI, 1.03–1.45), but a 3.6-fold increase in the risk of treatment-related death (RR: 3.5; 95% CI, 1.8–6.7; ref. 59).

A later randomized phase III trial compared paclitaxel alone versus paclitaxel and carboplatin in advanced NSCLC and found no statistically significant improvement in overall survival (OS) in the combined therapy (57). The 1-year survival was 32% and 37% for single and combination therapy, respectively, with a HR of 0.91 (95% CI, 0.77–1.17; P = 0.25; ref. 57). Hematologic toxicity and nausea were more frequent in the combination arm, but febrile neutropenia and toxic deaths were equally low in both arms.

A more recent meta-analysis comparing combined chemotherapy as second-line therapy with single-agent chemotherapy in the treatment of patients with advanced NSCLC from phase II and III clinical trials found no significant difference in OS between the combined and single-agent therapy (P = 0.32). However, doublet chemotherapy caused more grade 3 to 4 hematologic (41% vs. 25%; P = 0.0001) and nonhematologic toxicities (28% vs. 22%; P = 0.034; ref. 67).

In breast cancer, a meta-analysis of 43 randomized controlled trials included 9,742 patients who received chemotherapy in combination or a single agent as their first-line of treatment (60). They found that combined therapy significantly improved OS and time to progression (HR, 0.88, 95% CI, 0.83–0.93, P < 0.00001) compared with single-agent therapy but only delayed time to progression by a matter of months (60). Furthermore, women who received combined therapy faced serious side effects, such as vomiting and nausea, due to the toxicity of the treatment (60). In general, combining chemotherapy treatments showed a notable correlation with higher rates of leukopenia (OR 1.45; 95% CI, 1.28–1.65; P < 0.00001), alopecia (OR 1.55; 95% CI, 1.32–1.81; P < 0.00001), and nausea and vomiting (OR 1.65; 95% CI, 1.41–1.93; P < 0.00001), compared with single-agent treatment (60).

A meta-analysis of first-line chemotherapy in advanced gastric cancer on randomized phase II and III trials found that combined drug therapy (two or three drugs) improved survival (HR, 0.83; 95% CI, 0.74–0.93) compared with single-drug therapy. However, toxicities related to the treatment were higher in combined strategies (68). A study comparing the combination of three drugs (docetaxel, cisplatin, and fluorouracil or DCF) versus two drugs (cisplatin and fluorouracilor or CF) in phase III as first-line therapy of advanced gastric cancer found that OS (P = 0.02) and time to progression (P < 0.001) were longer for three drugs compared with two drugs. In addition, the 2-year survival rate on three drugs was 18% versus 9% for two drugs, led to higher toxicity (69) and clearly did not solve the problem of therapeutic resistance. However, grade 3 to 4 treatment-related adverse events were more frequent in the DCF group (69% vs. 59% in the CF group), particularly complicated neutropenia (29% in DCF vs. 12% in CF; ref. 69).

Meta-analyses of phase II and III clinical trials in pancreatic cancer have shown an advantage in using multidrug therapies, with improvements in tumor response and patient survival, but cures were still rare, and combination therapies increased toxicities compared with single-drug therapies. Heinemann and colleagues (63) analyzed patients with advanced or metastatic pancreatic cancer and found that combining gemcitabine along with another cytotoxic drug versus gemcitabine alone produced a small but significant improvement in OS (HR: 0.91; 95% CI, 0.85–0.97; P = 0.004). However, there is significantly more grade 3 to 4 toxicity in the combined regimens (70). This may help explain why patients with good performance status (PS) had a notable survival advantage with combination chemotherapy (HR, 0.76; P < 0.0001), whereas those with poor PS did not benefit (HR, 1.08; P = 0.40; ref. 63).

Another study by Moore and colleagues (71) on 569 patients of randomized phase III trials with advanced pancreatic cancer showed both OS (HR, 0.82; P = 0.083; 95% CI, 0.69–0.99) and 1-year survival (23% vs. 17%, P = 0.023) improvements using a combination of gemcitabine with erlotinib compared with using gemcitabine alone. Progression-free survival was also significantly better in this combination compared with single-agent therapy (HR, 0.77; 95% CI, 0.64–0.92; P = 0.004). However, there were few cures, and adding erlotinib to gemcitabine was associated with more grade 1 or 2 toxicity (71).

Pusceddu and colleagues (72) conducted a meta-analysis comparing the efficacy of two chemotherapy regimens in treating patients with metastatic pancreatic cancer: gemcitabine with nab-paclitaxel versus FOLFIRINOX (5-fluorouracil, oxaliplatin, and irinotecan). Neither multidrug therapy reliably achieved cures, and there was no difference in the OS between the 2- and 3-drug regimens (HR, 0.99; P = 0.9; 95% CI, 0.84–1.16). Both regimens were associated with grade 3 and 4 toxicities, with more anemia and neurotoxicity in gemcitabine with nab-paclitaxel and more neutropenia in FOLFIRINOX (72).

A meta-analysis of four randomized II and III trials with a total of 1,300 patients with ovarian cancer comparing single-agent platinum chemotherapy versus platinum combined with another chemotherapy (e.g., carboplatin with gemcitabine), in women with relapsed platinum-sensitive ovarian cancer, found an improvement in OS when using combined platinum chemotherapy (HR, 0.80; 95% CI, 0.64–1.00; P = 0.05; ref. 65). Two of the four trials included quality of life data, which showed no adverse effect on the quality of life between the single and combined platinum treatments (65). The 3-year OS when using carboplatin alone was 29% compared with 42% when using both carboplatin and epidoxorubicin (OR = 0.8; 95% CI, 0.6–1.2). However, platinum combined therapy led to higher toxicity (65).

Multidrug therapies that include immunotherapies and/or targeted therapies also tend to fail in late-stage cancers, showing that acquired therapeutic resistance is not just a problem for chemotherapies. Combining immune checkpoint inhibitors has shown significant improvements in OS across various malignancies, yet long-lasting survival is achieved by only 20% to 40% of patients (73), and increased toxicity is common (7476). Dual immune checkpoint blockade with anti–PD-1 and anti–CTLA-4 has resulted in high response rates (58%) in patients with metastatic melanoma, but nearly half of them experienced severe treatment-related side effects, with uncertain long-term survival benefits (75, 76).

Adding an immune checkpoint inhibitor to chemotherapy has shown some evidence of minor improvements in OS but also increased toxicity and little evidence of cures in triple-negative breast cancer (77, 78) and head and neck squamous cell carcinoma (79), as well as gastric, gastro-esophageal, and esophageal cancer (8082). Similarly, adding targeted therapy to chemotherapy produced a minor improvement in survival along with an increase in grade 3 to 4 toxicity in a meta-analysis of gastric cancer (83), although it did not improve OS in meta-analyses of NSCLC (84) or triple-negative breast cancer (85).

In all these studies and meta-analyses, investigators found evidence of increased efficacy for multiple drugs over single drugs, often at the cost of increased toxicity and treatment-related deaths. However, we must face the fact that whereas multidrug therapy may extend life by months, it rarely achieves a cure in late-stage disease, and there are diminishing returns from adding more drugs due to increases in toxicity and the evolution of multidrug resistance. In other words, multidrug therapy does not solve the problems of cancer or pest infestations.

Insects have evolved resistance to pesticides through a variety of mechanisms that are familiar to cancer biologists. These include modifications to the drug target, metabolic detoxification, reduced activation of the drug, prevention of drug uptake, and cellular efflux pumps. Insects often evolve resistance through changes in the structure of the target molecule, called “target site insensitivity.” These mutations generally reduce the binding affinity of the drug for the target molecule (8689). Drug target modifications are often observed in cancer, including the well-known T790M mutation in EGFR accounting for more than 50% to 60% of the cases of resistance to erlotinib therapy in lung adenocarcinoma (90, 91). Both pests and cancer cells can evolve resistance through amplification or overexpression of the target gene (92, 93). Another common mechanism of resistance to pesticides is metabolic detoxification through cytochrome P450s, esterases, and glutathione S-transferases (88), which have also been observed in cancers (94, 95). In some cases, resistance to a drug or a prodrug may evolve via inactivation or downregulation of the enzyme that would have activated the drug. This is seen in both proinsecticide resistance (9698) and cancer prodrug resistance (86, 87). There is (less common) evidence of resistance through prevention of uptake of drugs by cancer cells (99, 100) and pests (101, 102). Finally, cellular efflux pumps, such as major facilitator superfamily and ATP binding cassette transporters, are often implicated in pesticide resistance (101, 103105) as well as cancer drug resistance (86, 87, 99, 106) and in both cases can generate multidrug resistance, including cross-resistance between chemotherapies, radiotherapy, and immunotherapy (107). Like cancer, in some cases, pesticides trigger upregulation of resistance phenotypes like detoxification enzymes and efflux pumps, but can also be mutagens, causing de novo resistance mutations (108).

Some mechanisms of resistance seem to be unique to either pest or cancer cell resistance. In some cases, insects evolve resistance to pesticides through behavioral or physiologic changes, such as changes to the cuticle that prevent penetration of the drug or excretion of the drug from the insect’s body, that do not have a direct parallel in cancer cells (109, 110). Conversely, cancers often evolve resistance to drugs through activating pathways that compensate for a drug’s effect, such as an alternative proliferative signal, DNA repair to compensate for DNA damaging drugs (90, 91, 111, 112), or evolving alternative methods of evading the immune system under immunotherapy (113117). Cancers also disable apoptotic pathways and thereby prevent drug-induced cell death (99, 100, 111, 118). Cancers have also been shown to upregulate a gene (MUC4) that binds to the target of a drug (HER2) and out-competes the drug (trastuzumab) while activating the HER2 receptor (30). The absence of reports of these forms of drug resistance for pesticides may be due to the fact that most pesticides do not work through shutting down a proliferation signaling pathway or triggering apoptosis but achieve lethality through other mechanisms such as neurotoxicity (92). There are exceptions. Methoprene and methoxyfenozide seem to kill insects through inhibition of cellular proliferation (119), and azadirachtin A induces cell-cycle arrest and apoptosis through p53 activation (120); however, pest resistance seems to evolve through detoxification, not pathway compensation or disabling apoptosis (121124).

Through decades of experimentation and observation, pest managers have learned to focus on management, rather than eradication of pests (13). They have developed a set of principles for maintaining long-term management of pest populations (Fig. 1A; refs. 8, 19):

  • Prevention and suppression: adjust crops and the habitat to suppress the proliferation of pests and favor crop production.

  • Monitor continuously using systems for early warnings.

  • Decision-making: identify acceptable thresholds of infestation below which there will be no intervention and change interventions in response to changes in pest levels.

  • Prefer nonchemical control methods including mechanical, biological, and habitat controls.

  • Use pesticides as specific as possible to the type of pest, minimizing off-target effects, including pesticides that modify pest behavior rather than kill the pests.

  • Use the lowest effective dose possible.

  • Choose pesticides to reduce cross-resistance and separate their application in space and time.

  • Evaluate success based on long-term management.

  • Forecast pest growth and response to interventions to inform most of the other principles.

Figure 1.

Lessons from IPM to oncology. A, These are the best practices in pest management and how they might be applied in cancer treatment. B, Comparison of standard therapy with two models of adaptive therapy. In standard therapy (A), the MTD of a drug is used. In dose modulation adaptive therapy (B), the dose of the drugs is adjusted based on the tumor’s response. In drug holiday (C) treatment skipping, the MTD of the drug is used until the tumor shrinks and then the treatment is skipped. CAR, chimeric antigen receptor.

Figure 1.

Lessons from IPM to oncology. A, These are the best practices in pest management and how they might be applied in cancer treatment. B, Comparison of standard therapy with two models of adaptive therapy. In standard therapy (A), the MTD of a drug is used. In dose modulation adaptive therapy (B), the dose of the drugs is adjusted based on the tumor’s response. In drug holiday (C) treatment skipping, the MTD of the drug is used until the tumor shrinks and then the treatment is skipped. CAR, chimeric antigen receptor.

Close modal

Pest managers are aware that no single intervention is sufficient to control pests, and that usually the use of several chemicals is necessary (8). However, combining these principles and combining chemical controls with other forms of controls can lead to the effective and long-term management of pests (8, 14, 15). Hence, in the term integrated pest management, the emphasis is on both integration and management.

The principles of IPM may be applied to any type of cancer. Here, we describe how these principles might be employed in the design of a clinical trial for colorectal cancer as a concrete example to illustrate these ideas. Of course, before such a trial is opened, the innovations from IPM that have not yet been tested in animal models should be studied in preclinical models. We chose colorectal cancer because (i) it is common (125); (ii) it is difficult to manage the late stages of this disease (126); and (iii) there is a variety of drugs that can produce an initial response (127), and so could be rotated, or adjusted in an IPM-inspired protocol. Inclusion criteria for such a clinical trial should prioritize stage III and IV patients that have not received prior chemotherapeutic interventions, or for which only one MoA has failed and where complete resection is either not possible or was unsuccessful. The primary outcome from this study should be OS, with secondary outcomes including measures of quality of life and time to progression (defined as the tumor continuing to grow despite the use of all available drugs). The goal here is to manage cancer as a chronic disease so that patients can live with their cancers but not die from them, or from the treatments.

Prevention and suppression

Cancer biologists, like pest managers, have noted that prevention should be a key part in the strategy of dealing with cancer (1, 3, 10, 128), well before the point of a therapeutic clinical trial. It is much easier to prevent an infestation or cancer than it is to treat it. Translating these ideas to oncology might involve modifying the microenvironment of a benign tumor to favor healthy cells and disfavor malignant cells (129), which we discuss below under habitat controls. Other strategies, such as improving the overall health of patients through diet and exercise, and other lifestyle factors are familiar and could be helpful in making the tissue microenvironments less favorable for cancer (130).

Decision-making (consider the patient’s biology, drug, and target cells)

An ideal trial based on IPM should start with extensive sampling and profiling the patient’s cancer to determine what known forms of resistance are present in the tumor as well as what targetable mutations are present and at what frequencies, as well as whether the tumor is growing. Pest managers would not even treat a field if the infestation is below acceptable levels of crop destruction. Translated to oncology, this would suggest watchful waiting as long as the tumor is small enough and there are no signs of significant damage to the patient. The principle is to establish a threshold below which we do not treat. These thresholds may be based on multiple objectives such as quality of life, risk of progression, PS, and other measures of health. We do not currently know what the tradeoffs will be between starting therapy early and selecting for resistance early versus living with some low tumor burden and the potential damage from that (131). The specific thresholds to be used will have to be determined by theoretical, preclinical, and clinical experiments. Withholding therapy does not imply missing the opportunity to cure the patient. Rather, withholding therapy preserves the opportunity to use drugs effectively in the future to control the tumor.

Decision-making should take into account and exploit aspects of the pest (and cancer cell) biology. For example, Lygus bugs, which damage cotton, lay their eggs in the height of summer. They take 1 week to hatch into nymphs and another week to develop to the point that they begin to damage crops. Efficacy of control is optimized by timing and targeting for nymphs. Pest managers exploit this synchronized development with a 2-week window to detect and treat the crops. Similarly, cancer therapy efficacy can be enhanced through careful timing of drug application, either by timing chemotherapy to circadian cycles so as to protect normal tissues (132) or by attempts to synchronize cancer cells in order to sensitize them to therapy (133135).

Monitor regularly and change interventions in response

It is unclear how often a tumor should be monitored during treatment as this will vary between patients, tumor types, and interventions. It may even vary temporally within a given patient as the dynamics of their cancer slow down or speed up. Traditionally, oncologists do not monitor tumor burden during a therapy protocol and only check weeks to months later to determine whether the therapy is effective. For example, in a clinical study that was conducted on patients with rectal adenocarcinoma, their response to chemoradiation was determined only 2 to 4 weeks after the treatment was completed (136). In cases in which a solid tumor is monitored during therapy, RECIST guidelines are to evaluate it every 6 to 8 weeks (137), although evaluation every 12 weeks is common (138). Monitoring tumor response intensively during treatment allows for adjusting to its response.

Adaptive therapy is one example of the application of pest management to oncology, based on monitoring and adjusting therapy in response to tumor dynamics. Adaptive therapy seeks to prevent the expansion of therapeutically resistant clones by maintaining chemosensitive cells in the tumors to compete with the resistant cells (45, 46, 139, 140). This is done operationally by trying to keep the tumor at a stable size. In the dose modulation version of adaptive therapy, the tumor burden is measured frequently and the dose is increased if the tumor is growing, but the dose is lowered if it is shrinking. Otherwise, if the tumor is stable, dosing is kept at the same level (Fig. 1B). This dramatically extended time to progression and survival in mouse models of ovarian and breast cancer (45, 139, 141). The amount of drug required to keep the tumor stable also decreased over time, possibly because tumors in the adaptive therapy arms evolved better perfusion than the tumors in the MTD arms (45, 139). The only clinical trial of adaptive therapy that has completed to date used a different protocol, similar to intermittent therapy, in which dosing stopped when the tumor dropped below 50% of its initial burden, and was restarted when it recovered to the level of the initial burden. This resulted in the doubling of radiographic progression-free survival in castration-resistant metastatic prostate cancer (47). Dose adjustment works better than intermittent adaptive therapy in both computational and mouse models (45, 139, 142). In a colorectal cancer clinical trial, we would propose using dose modulation adaptive therapy with each single drug until the tumor grows and then rotating to a new drug with a different MoA.

Prefer nonchemical control methods

One way to prevent the evolution of therapeutic resistance to drugs is to use nonchemical forms of control. Pest managers distinguish three types of nonchemical controls: mechanical, habitat, and biological.

Mechanical controls

Pest burden can be reduced by physically removing pests, erecting barriers to their entry, and setting up traps to contain them. Using “lure and kill” traps has been shown to be an effective supplemental strategy in a variety of pest management studies (143). Surgery is a common technique for physically removing cancer cells (144). Removing the primary tumor and metastases reduces the size of the cancer cell population and thereby reduces the evolvability of cancer. This should be beneficial even if not all metastases can be removed (145). In fact, mechanical removal of tumors (cytoreductive surgery) has been shown to increase survival in many cases of metastatic cancer, where the tumor burden can be substantially reduced (145, 146). This was true even for recurrent cancer (147). However, the benefits must be weighed against the morbidity and risks of the surgery. Cancer traps, potentially including chemoattractants for cancer cells, might complement other control mechanisms and help prevent further metastases (148, 149).

Habitat controls

Translated to oncology, using habitat controls would mean making the microenvironment unfavorable to the cancer cells and favorable to the normal cells as much as possible. In some cases, we might be able to exploit the loss of functions that may accumulate in cancers. Fasting may cause normal cells to become quiescent because they respond to growth inhibition signals whereas cancer cells likely continue to proliferate. This can protect normal cells from the toxic effects of cell cycle–specific drugs (150). Antiangiogenic drugs are designed to make the microenvironment less hospitable for cancers, although they have had mixed success (151).

Biological controls

Biological control in cancer treatment includes immunotherapy (predators) as well as viral and bacterial therapy. Engaging predation from the immune system may include cancer vaccines (152), immune checkpoint modulators (153), and chimeric antigen receptor T-cell therapy (154, 155).

Use drugs with specificity

Using drugs specific to a cancer helps to minimize off-target effects. Cancer cells are derived from human cells, so it is inherently difficult to develop cancer-specific therapies that do not affect normal cells. If sequencing or other profiling of cancer identifies clonal mutations that may be targeted by a drug, they should be included in the rotation of therapies along with the more broad-spectrum therapies.

Use the lowest dose possible

Most chemotherapeutic drugs can be characterized by a dose–response curve; the higher the drug dose delivered, the more cancer cells are killed (156, 157). This relationship between dose and cell death is limited at high doses by diminishing returns as well as unacceptable toxicity (156, 157). This relationship naturally led to the use of the MTD in most cancer therapy protocols (158, 159). For drugs for which toxicity is not limiting, like some hormone and targeted therapies, researchers have used the minimum dose that achieves the maximum biological efficacy, called the optimal biological dose (OBD; ref. 160). There is no consensus as to how “efficacy” should be defined in the OBD. If it is defined by tumor shrinkage, then this will lead to very high selective pressures on the cancer cells (160, 161). Unfortunately, we know from evolutionary theory that using the maximum tolerated (or effective) dose is also the fastest way to select for acquired therapeutic resistance (140). The strength of selection is directly related to the speed of evolution. This implies that we should be using the minimum dose necessary to control the tumor in order to minimize the selective pressure for resistance. Simulation studies bear this out (142). Dose modulation adaptive therapy is a protocol for finding that dose (45, 46, 139, 140). Note that if efficacy is defined as OS time of the patients, then the OBD may well be something close to the minimum dose necessary to control the tumor.

Reduce cross-resistance

Pest managers have learned to reduce cross-resistance by diversifying the MoA of drugs they use and partitioning those drugs in space and time as much as possible (8, 1315).

Diversify the use of MoAs as much as possible

Cancer drugs may be grouped by the MoA they use to achieve their anticancer effects. Both theory and experiment show that resistance to one drug with a particular MoA also confers cross-resistance to other drugs that use the same MoA (162, 163). Thus, to avoid cross-resistance and reap the benefits of using multiple drugs, those drugs should be chosen so that they use different MoAs (19). This principle has been recognized in oncology, although it is not always followed (164, 165). We might be able to do even better in improving patients’ survival by combining drugs for which resistance to one drug causes sensitivity to the other, and vice versa, an approach called “double-bind therapy” in cancer (19, 128, 166, 167) and “negative cross-resistance” in pest management, in which cytochrome P450s detoxify some pesticides but activate other pesticide prodrugs (168).

Partition MoAs in space or time to segregate their usage

As we discussed above, the combination of drugs with different MoAs does not solve the problem of acquired therapeutic resistance, due to phenomena like multidrug resistance in both cancer (106) and pest management (169, 170). So pest managers advise separating the application of different drugs as much as possible in both time and space, including refuges that are not sprayed so as to preserve drug-sensitive pests (14). Although spatial partitioning is difficult in cancer, drugs with rapid half-lives could be used so that one drug is completely cleared from the system before cancer cells are exposed to the next drug (171).

Perhaps the most striking difference between oncology and pest management is that pest managers try to never apply the same MoA twice in a row, and never more than twice within a growing season (e.g., the Arizona Cotton IPM strategy; Fig. 2A). In oncology, we repeatedly apply the same drug or drug combinations, week after week. In contrast, pest managers endeavor to switch pesticides every time they spray a field (8). This prevents a resistant clone from expanding much before it is exposed to a new drug with a different MoA. This heuristic of only applying a drug once, or at most twice, leads to a different perspective on decision-making compared with oncology. Pest managers will sometimes delay the application of a drug to maximize its impact if they anticipate a coming influx of pests, even if the current pest level is above the threshold that would normally trigger treatment. This difference from traditional protocols in oncology derives from the goal of control, rather than cure.

Figure 2.

Schematic of a MoA schedule. A, In pest management, pest managers avoid sequential use of the same MoA and avoid using a MoA more than twice within the same growing season. To avoid cancer cells acquiring therapeutic resistance, repeated applications of a MoA should be separated as much as possible. B, Proposed scheduling of drugs that applies each drug once per 4-week cycle and uses different MoAs. The third week of each cycle uses an antiangiogenic drug to avoid restricting the vasculature before trying to deliver the other drugs. Week 4 of each cycle is a drug holiday break to allow the patient to recover.

Figure 2.

Schematic of a MoA schedule. A, In pest management, pest managers avoid sequential use of the same MoA and avoid using a MoA more than twice within the same growing season. To avoid cancer cells acquiring therapeutic resistance, repeated applications of a MoA should be separated as much as possible. B, Proposed scheduling of drugs that applies each drug once per 4-week cycle and uses different MoAs. The third week of each cycle uses an antiangiogenic drug to avoid restricting the vasculature before trying to deliver the other drugs. Week 4 of each cycle is a drug holiday break to allow the patient to recover.

Close modal

We identified existing FDA-approved drugs for colorectal cancer with four distinct MoAs: DNA damaging agents, including DNA synthesis blockades and platinum therapies, antimetabolites, EGFR inhibitors, and VEGF inhibitors. After 3 weeks (with one drug each week), the patients would receive a further week break to complete the 4-week cycle. The third week in each cycle could be an antiangiogenic drug (to restrict the tumor vasculature and nutrients once the other drugs have been delivered; Fig. 2B). This restriction of resources should make it especially hard for resistant cells to grow in comparison to sensitive cells, because the former often require more resources to pay the fitness cost of resistance (45).

Evaluation based on long-term control

Prior to the development of IPM, farmers would generally evaluate pest management methods by a combination of crop yields and the absence of pests (8). Practices prior to IPM were usually “womb-to-tomb” spraying, in which pesticides were used until the end of the production cycle (172). Successful management of pest resistance required a shift in evaluation criteria away from short-term response and the impractical goal of total pest eradication to the more helpful goals of long-term management, cumulative yield, and stability of yield over time (8). The parallel for oncologists may be to move away from partial and complete responses as the criteria of evaluation and focus on time to progression, survival time, and quality of life.

Forecasting

Forecasting is central to IPM efforts and it informs much of what pest managers do including decision-making, monitoring, prevention efforts, managing resistance, and evaluation. However, formal forecasting is rarely applied in oncology. Gu and colleagues (173) studied patients with glioblastoma multiforme and demonstrated the impact of a predictive model based on patient-specific planning of the treatment strategy. It is helpful to fit models of sensitive and resistant cell dynamics to tumor burden measures in order to forecast the expected results of continuing the current treatment versus changing treatment (174, 175). The more often a tumor can be monitored, the better those forecasts can be (176).

Summary of the clinical trial

Each of the innovations in this example clinical trial ought to first be evaluated in preclinical models (141), although those models must demonstrate acquired therapeutic resistance under the standard of care in order to faithfully represent the clinical challenge. Colorectal cancer remains an appealing candidate disease due to the existence of well-described spontaneous murine models (177). If preclinical experiments with the above design are successful, clinical trials may be able to bypass phase I clinical trials due to the fact that safe dosage ranges for the drugs in our design would have already been determined. For phases II and III, patient recruitment should involve those in the second line of treatment and exclude drugs with a similar MoA from the first line of treatment that was used for those patients.

Challenges and clinical needs for IPM-inspired oncology

In order to develop IPM-inspired treatments for cancer, there are a few important challenges that will need to be addressed. First, it is not clear how frequently we will need to measure that tumor burden to appropriately adjust our interventions. We need economical and safe ways to measure tumor burden in order to monitor cancers frequently. This may require the development of new technologies.

Liquid biopsies (blood, serum, or plasma, as well as urine, cerebrospinal fluid, saliva, and pleural effusion fluids) carry a wealth of information that might be used to monitor tumor burden and response, including circulating tumor cells, ctDNA, circulating tumor RNA (particularly microRNAs), tumor-educated platelets (178, 179), extracellular vesicles (180), and proteins (181, 182). ctDNA in the blood is particularly promising as a minimally invasive biomarker for monitoring cancer (183). It may not only measure tumor burden but also be more generally representative of the cancer cell population and molecular diagnosis (mutations of the tumor) than a single biopsy (183). It may also reveal mutations that cause resistance and thus enable monitoring of some resistant populations as well as overall tumor burden (184186). ctDNA has been shown to be clinically useful across a variety of domains including diagnosis and early detection, monitoring treatment response, detecting minimal residual disease as well as recurrence, and generally guiding personalized therapy (187).

There are a variety of different assays applied to ctDNA, from targeted panels with high sensitivity (188, 189) to genome wide assays (190) and to methylation (191, 192) or fragment patterns (193), which show promise. However, methods for using ctDNA as a tumor marker are still under development and are not yet standardized for most clinical applications (181, 182, 184, 187, 193195).

Second, we need biomarkers to help predict if a cancer is curable or if we should focus on management instead. In particular, if we could distinguish cancers that are likely to harbor therapeutically resistant clones from cancers that do not, perhaps through measuring intratumor heterogeneity (196), we could focus on controlling the former and curing the latter.

Finally, many of the ideas coming from IPM would need to be tested in animal models to provide sufficient evidence for their efficacy to justify clinical trials.

Drug resistance is one of the most important problems we face in clinical oncology. Solving it will likely require the integration of oncology with evolutionary biology, ecology, bioinformatics, and inspiration from other fields involved in managing systems that evolve therapeutic resistance, including pest management, weed management, and infectious diseases (viruses, bacteria, protozoa, fungi, and helminths; refs. 197, 198).

Here, we have shown that pest managers face the same problem as oncologists, with pesticides selecting for resistant pests. In both cases, a single drug tends to fail. Combination therapy with different MoAs is more effective than single-drug therapies but still leads to multidrug resistance after a short period of time. However, IPM dynamically slows the development of pesticide-resistant pests often below densities requiring control over the long term (15). Nonchemical and chemical approaches used in IPM may be translated to cancer therapy. Modern principles of resistance management could control the growth of resistance by limiting the use of each MoA (minimizing the frequency of use and dose to the lowest practical levels), diversifying the use of MoAs through rotation programs, partitioning MoAs use in space or time, and utilizing nonchemical methods of control.

In fact, a general principle underlying most of the lessons of IPM is that the more drugs we use, the more we select for resistance. Work on therapeutic resistance in malaria has come to similar conclusions (199, 200). Most strategies of IPM are ways to limit the amount of drug used, although still controlling the infestation. IPM inspired adaptive therapy in oncology (45, 139, 140). An ideal trial based on IPM would start with intensively monitoring cancer and rotating drugs at minimum effect doses, stopping dosing when possible and avoiding using any one MoA for so long as to select for resistance. If we assume that resistant cells are already present, as pest managers do, this approach represents a fundamental shift from trying to cure cancer to controlling it, with the aim of prolonging patient survival and quality of life. This is a call to carry out the preclinical experiments and clinical trials to test these ideas and generate the evidence for improving clinical practice. By transcending the sole focus on a cure, we open up new possibilities for methods to dramatically improve both patient survival and quality of life.

S. Seyedi reports other support from Antharis Therapeutics outside the submitted work. Z. Compton reports grants from NIH during the conduct of the study. P.C. Ellsworth reports grants from USDA-CPPM-EIP during the conduct of the study, as well as grants, nonfinancial support, and other support from various agricultural input suppliers outside the submitted work. No disclosures were reported by the other authors.

The authors thank Gabrielle Hirnese for help with the figures. This work was supported in part by NIH grants T32 CA272303, U54 CA217376, U2C CA233254, P01 CA91955, and R01 CA140657 as well as CDMRP Breast Cancer Research Program Award BC132057 and the Arizona Biomedical Research Commission grant ADHS18-198847. The findings, opinions, and recommendations expressed here are those of the authors and not necessarily those of the universities where the research was performed or the NIH. This material is also based on work that is supported by the National Institute of Food and Agriculture, US Department of Agriculture, under award number 2021-70006-35385.

1.
Fortunato
A
,
Boddy
A
,
Mallo
D
,
Aktipis
A
,
Maley
CC
,
Pepper
JW
.
Natural selection in cancer biology: from molecular snowflakes to trait hallmarks
. In:
Swanton
C
,
Bardelli
A
,
Polyak
K
,
Shah
S
,
Graham
T
, editors.
Cancer evolution
.
Cold Spring Harbor (NY)
:
Cold Spring Harbor Laboratory Press
;
2016
.
2.
Andor
N
,
Graham
TA
,
Jansen
M
,
Xia
LC
,
Aktipis
CA
,
Petritsch
C
, et al
.
Pan-cancer analysis of the extent and consequences of intratumor heterogeneity
.
Nat Med
2016
;
22
:
105
13
.
3.
Schiffman
JD
,
White
RM
,
Graham
TA
,
Huang
Q
,
Aktipis
A
.
The darwinian dynamics of motility and metastasis
. In:
Maley
CC
,
Greaves
M
, editors.
Frontiers in cancer research: evolutionary foundations, revolutionary directions
.
New York (NY)
:
Springer
;
2016
. p.
135
76
.
4.
Casás-Selves
M
,
Degregori
J
.
How cancer shapes evolution, and how evolution shapes cancer
.
Evolution (N Y)
2011
;
4
:
624
34
.
5.
Tabashnik
BE
.
Managing resistance with multiple pesticide tactics: theory, evidence, and recommendations
.
J Econ Entomol
1989
;
82
:
1263
9
.
6.
Wang
X
,
Zhang
H
,
Chen
X
.
Drug resistance and combating drug resistance in cancer
.
Cancer Drug Resist
2019
;
2
:
141
60
.
7.
Chabner
BA
,
Roberts
TG
Jr
.
Timeline: chemotherapy and the war on cancer
.
Nat Rev Cancer
2005
;
5
:
65
72
.
8.
Barzman
M
,
Bàrberi
P
,
Birch
ANE
,
Boonekamp
PM
,
Dachbrodt-Saaydeh
S
,
Graf
B
, et al
.
Eight principles of integrated pest management
.
Agron Sustain Dev
2015
;
35
:
1199
215
.
9.
Mendes
KF
,
De Sousa
R
,
Mielke
KC
.
Biodegradation technology of organic and inorganic pollutants
.
London
:
InTechOpen Limited
;
2022
.
10.
Gould
F
,
Brown
ZS
,
Kuzma
J
.
Wicked evolution: can we address the sociobiological dilemma of pesticide resistance?
Science
2018
;
360
:
728
32
.
11.
Lin
A
,
Giuliano
CJ
,
Palladino
A
,
John
KM
,
Abramowicz
C
,
Yuan
ML
, et al
.
Off-target toxicity is a common mechanism of action of cancer drugs undergoing clinical trials
.
Sci Transl Med
2019
;
11
:
eaaw8412
.
12.
Lamichhane
JR
,
Aubertot
J-N
,
Begg
G
,
Birch
ANE
,
Boonekamp
P
,
Dachbrodt-Saaydeh
S
, et al
.
Networking of integrated pest management: a powerful approach to address common challenges in agriculture
.
Crop Prot
2016
;
89
:
139
51
.
13.
Stenberg
JA
.
A conceptual framework for integrated pest management
.
Trends Plant Sci
2017
;
22
:
759
69
.
14.
Naranjo
SE
,
Ellsworth
PC
.
Landscape considerations in pest man- agement: case study of the Arizona cotton IPM system
. In:
Brewer
MJ
,
Hein
GL
, editors.
Arthropod management and landscape considerations in large-scale agroecosystems
.
Oxfordshire (UK)
:
CABI
;
2024
. p.
44
77
.
15.
Ellsworth
PC
,
Fournier
A
.
Theory versus practice: are insecticide mixtures in Arizona cotton used for resistance management?
Pest Manag Sci
2024 Jun 25
[
Online ahead of print
].
16.
Stern
VM
,
Smith
RF
,
Van den Bosch
R
,
Hagen
KS
.
The integration of chemical and biological control of the spotted alfalfa aphid: the integrated control concept
.
Hilgardia
1959
;
29
:
81
101
.
17.
Chakraborty
S
,
Rahman
T
.
The difficulties in cancer treatment
.
Ecancermedicalscience
2012
;
6
:
ed16
.
18.
Qian
C-N
,
Mei
Y
,
Zhang
J
.
Cancer metastasis: issues and challenges
.
Chin J Cancer
2017
;
36
:
38
.
19.
Cunningham
JJ
.
A call for integrated metastatic management
.
Nat Ecol Evol
2019
;
3
:
996
8
.
20.
Parkin
EA
,
Scott
EIC
,
Forester
R
.
Increase resistance of stored-product insects to insecticides. The resistance field strains of beetles (C) Tribolium castaneum
.
Pest Infest Res
1962
;
21
:
34
5
.
21.
Ballantyne
GH
,
Harrison
RA
.
Genetic and biochemical comparisons of organophosphate resistance between strains of spider mites (Tetranychus species: Acari)
.
Entomol Exp Appl
1967
;
10
:
231
9
.
22.
Champ
BR
,
Campbell-Brown
MJ
.
Insecticide resistance in Australian Tribolium castaneum (herbst) (Coleoptera, Tenebrionidae)—II: malathion resistance in eastern Australia
.
J Stored Prod Res
1970
;
6
:
111
31
.
23.
Onstad
DW
,
Carrière
Y
.
The role of landscapes in insect resistance management
. In:
Insect resistance management
. 2nd ed.
Cambridge, (MA)
:
Academic Press
;
2014
. p.
327
71
.
24.
Levine
E
,
Oloumi-Sadeghi
H
.
Western corn rootworm (Coleoptera: Chrysomelidae) larval injury to corn grown for seed production following soybeans grown for seed production
.
J Econ Entomol
1996
;
89
:
1010
6
.
25.
Goodman
LS
,
Wintrobe
MM
.
Nitrogen mustard therapy; use of methyl-bis (beta-chloroethyl) amine hydrochloride and tris (beta-chloroethyl) amine hydrochloride for Hodgkin’s disease, lymphosarcoma, leukemia and certain allied and miscellaneous disorders
.
J Am Med Assoc
1946
;
132
:
126
32
.
26.
Farber
S
,
Diamond
LK
,
Mercer
RD
,
Sylvester
RF
,
Wolff
JA
.
Temporary remissions in acute leukemia in children produced by folic acid antagonist, 4-aminopteroyl-glutamic acid (aminopterin)
.
N Engl J Med
1948
;
238
:
787
93
.
27.
Wu
Y
,
Song
Y
,
Wang
R
,
Wang
T
.
Molecular mechanisms of tumor resistance to radiotherapy
.
Mol Cancer
2023
;
22
:
96
.
28.
Vasan
N
,
Baselga
J
,
Hyman
DM
.
A view on drug resistance in cancer
.
Nature
2019
;
575
:
299
309
.
29.
Weisberg
E
,
Griffin
JD
.
Resistance to imatinib (Glivec): update on clinical mechanisms
.
Drug Resist Updat
2003
;
6
:
231
8
.
30.
Nahta
R
,
Esteva
FJ
.
HER2 therapy: molecular mechanisms of trastuzumab resistance
.
Breast Cancer Res
2006
;
8
:
215
.
31.
Seidman
AD
,
Fornier
MN
,
Esteva
FJ
,
Tan
L
,
Kaptain
S
,
Bach
A
, et al
.
Weekly trastuzumab and paclitaxel therapy for metastatic breast cancer with analysis of efficacy by HER2 immunophenotype and gene amplification
.
J Clin Oncol
2001
;
19
:
2587
95
.
32.
Kobayashi
S
,
Boggon
TJ
,
Tajhal
D
,
Jänne
PA
,
Olivier
K
,
Matthew
M
, et al
.
EGFR mutation and resistance of non–small-cell lung cancer to gefitinib
.
N Engl J Med
2005
;
352
:
786
92
.
33.
Tang
J
,
Salama
R
,
Gadgeel
SM
,
Sarkar
FH
,
Ahmad
A
.
Erlotinib resistance in lung cancer: current progress and future perspectives
.
Front Pharmacol
2013
;
4
:
15
.
34.
Soria
J-C
,
Ohe
Y
,
Vansteenkiste
J
,
Reungwetwattana
T
,
Chewaskulyong
B
,
Lee
KH
, et al
.
Osimertinib in untreated EGFR-mutated advanced non–small-cell lung cancer
.
N Engl J Med
2018
;
378
:
113
25
.
35.
Katayama
R
,
Shaw
AT
,
Khan
TM
,
Mino-Kenudson
M
,
Solomon
BJ
,
Halmos
B
, et al
.
Mechanisms of acquired crizotinib resistance in ALK-rearranged lung cancers
.
Sci Transl Med
2012
;
4
:
120ra17
.
36.
Ribas
A
,
Hamid
O
,
Daud
A
,
Hodi
FS
,
Wolchok
JD
,
Kefford
R
, et al
.
Association of pembrolizumab with tumor response and survival among patients with advanced melanoma
.
JAMA
2016
;
315
:
1600
9
.
37.
Eggermont
AMM
,
Chiarion-Sileni
V
,
Grob
J-J
,
Dummer
R
,
Wolchok
JD
,
Schmidt
H
, et al
.
Prolonged survival in stage III melanoma with ipilimumab adjuvant therapy
.
N Engl J Med
2016
;
375
:
1845
55
.
38.
Syn
NL
,
Teng
MWL
,
Mok
TSK
,
Soo
RA
.
De-novo and acquired resistance to immune checkpoint targeting
.
Lancet Oncol
2017
;
18
:
e731
41
.
39.
Jenkins
RW
,
Barbie
DA
,
Flaherty
KT
.
Mechanisms of resistance to immune checkpoint inhibitors
.
Br J Cancer
2018
;
118
:
9
16
.
40.
Larkin
J
,
Chiarion-Sileni
V
,
Gonzalez
R
,
Grob
J-J
,
Rutkowski
P
,
Lao
CD
, et al
.
Five-year survival with combined nivolumab and ipilimumab in advanced melanoma
.
N Engl J Med
2019
;
381
:
1535
46
.
41.
Topalian
SL
,
Hodi
FS
,
Brahmer
JR
,
Gettinger
SN
,
Smith
DC
,
McDermott
DF
, et al
.
Five-year survival and correlates among patients with advanced melanoma, renal cell carcinoma, or non–small cell lung cancer treated with nivolumab
.
JAMA Oncol
2019
;
5
:
1411
20
.
42.
Ozaki
K
.
Suppression of resistance through synergistic combinations with emphasis on planthoppers and leafhoppers infesting rice in Japan
. In:
Georghiou
GP
,
Saito
T
, editors.
Pest resistance to pesticides
.
Boston (MA)
:
Springer
;
1983
. pp.
595
613
.
43.
Pimentel
D
,
Burgess
M
.
Effects of single versus combinations of insecticides on the development of resistance
.
Environ Entomol
1985
;
14
:
582
9
.
44.
MacDonald
RS
,
Surgeoner
GA
,
Solomon
KR
,
Harris
CR
.
Effect of four spray regimes on the development of permethrin and dichlorvos resistance, in the laboratory, by the house fly (Diptera: Muscidae)
.
J Econ Entomol
1983
;
76
:
417
22
.
45.
Enriquez-Navas
PM
,
Kam
Y
,
Das
T
,
Hassan
S
,
Silva
A
,
Foroutan
P
, et al
.
Exploiting evolutionary principles to prolong tumor control in preclinical models of breast cancer
.
Sci Transl Med
2016
;
8
:
327ra24
.
46.
Gatenby
RA
.
A change of strategy in the war on cancer
.
Nature
2009
;
459
:
508
9
.
47.
Zhang
J
,
Cunningham
J
,
Brown
J
,
Gatenby
R
.
Evolution-based mathematical models significantly prolong response to abiraterone in metastatic castrate-resistant prostate cancer and identify strategies to further improve outcomes
.
Elife
2022
;
11
:
e76284
.
48.
Georghiou
GP
,
Lagunes
A
,
Baker
JD
.
Effect of insecticide rotations on evolution of resistance
. In:
Matsunaka
S
,
Hutson
DH
,
Murphy
SD
, editors.
Mode of action, metabolism and toxicology
.
Oxford (UK)
:
Pergamon
;
1983
. p.
183
9
.
49.
Bozic
I
,
Reiter
JG
,
Allen
B
,
Antal
T
,
Chatterjee
K
,
Shah
P
, et al
.
Evolutionary dynamics of cancer in response to targeted combination therapy
.
Elife
2013
;
2
:
e00747
.
50.
Delbaldo
C
,
Michiels
S
,
Syz
N
,
Soria
J-C
,
Le Chevalier
T
,
Pignon
J-P
.
Benefits of adding a drug to a single-agent or a 2-agent chemotherapy regimen in advanced non–small-cell lung cancer: a meta-analysis
.
JAMA
2004
;
292
:
470
84
.
51.
Sudo
M
,
Takahashi
D
,
Andow
DA
,
Suzuki
Y
,
Yamanaka
T
.
Optimal management strategy of insecticide resistance under various insect life histories: heterogeneous timing of selection and interpatch dispersal
.
Evol Appl
2017
;
11
:
271
83
.
52.
Madgwick
PG
,
Kanitz
R
.
Beyond redundant kill: a fundamental explanation of how insecticide mixtures work for resistance management
.
Pest Manag Sci
2023
;
79
:
495
506
.
53.
Djouaka
RJ
,
Atoyebi
SM
,
Tchigossou
GM
,
Riveron
JM
,
Irving
H
,
Akoton
R
, et al
.
Evidence of a multiple insecticide resistance in the malaria vector anopheles funestus in South West Nigeria
.
Malar J
2016
;
15
:
565
.
54.
Cohen
Y
,
Kosman
E
.
Procedures for calculating and differentiating synergism and antagonism in action of fungicide mixtures
.
Phytopathology
1996
;
86
:
1263
72
.
55.
Plana
D
,
Palmer
AC
,
Sorger
PK
.
Independent drug action in combination therapy: implications for precision oncology
.
Cancer Discov
2022
;
12
:
606
24
.
56.
Etzioni
R
,
Urban
N
,
Ramsey
S
,
McIntosh
M
,
Schwartz
S
,
Reid
B
, et al
.
The case for early detection
.
Nat Rev Cancer
2003
;
3
:
243
52
.
57.
Lilenbaum
RC
,
Herndon
JE
2nd
,
List
MA
,
Desch
C
,
Watson
DM
,
Miller
AA
, et al
.
Single-agent versus combination chemotherapy in advanced non-small-cell lung cancer: the Cancer and Leukemia Group B (study 9730)
.
J Clin Oncol
2005
;
23
:
190
6
.
58.
Bunn
PA
Jr
,
Kelly
K
.
New chemotherapeutic agents prolong survival and improve quality of life in non-small cell lung cancer: a review of the literature and future directions
.
Clin Cancer Res
1998
;
4
:
1087
100
.
59.
Lilenbaum
RC
,
Langenberg
P
,
Dickersin
K
.
Single agent versus combination chemotherapy in patients with advanced nonsmall cell lung carcinoma: a meta-analysis of response, toxicity, and survival
.
Cancer
1998
;
82
:
116
26
.
60.
Carrick
S
,
Parker
S
,
Thornton
CE
,
Ghersi
D
,
Simes
J
,
Wilcken
N
.
Single agent versus combination chemotherapy for metastatic breast cancer
.
Cochrane Database Syst Rev
2009
;
2009
:
CD003372
.
61.
Iacovelli
R
,
Pietrantonio
F
,
Maggi
C
,
de Braud
F
,
Di Bartolomeo
M
.
Combination or single-agent chemotherapy as adjuvant treatment of gastric cancer: a systematic review and meta-analysis of published trials
.
Crit Rev Oncol Hematol
2016
;
98
:
24
8
.
62.
Ciliberto
D
,
Botta
C
,
Correale
P
,
Rossi
M
,
Caraglia
M
,
Tassone
P
, et al
.
Role of gemcitabine-based combination therapy in the management of advanced pancreatic cancer: a meta-analysis of randomised trials
.
Eur J Cancer
2013
;
49
:
593
603
.
63.
Heinemann
V
,
Boeck
S
,
Hinke
A
,
Labianca
R
,
Louvet
C
.
Meta-analysis of randomized trials: evaluation of benefit from gemcitabine-based combination chemotherapy applied in advanced pancreatic cancer
.
BMC Cancer
2008
;
8
:
82
.
64.
Bria
E
,
Milella
M
,
Gelibter
A
,
Cuppone
F
,
Pino
MS
,
Ruggeri
EM
, et al
.
Gemcitabine-based combinations for inoperable pancreatic cancer: have we made real progress? A meta-analysis of 20 phase 3 trials
.
Cancer
2007
;
110
:
525
33
.
65.
Raja
FA
,
Counsell
N
,
Colombo
N
,
Pfisterer
J
,
du Bois
A
,
Parmar
MK
, et al
.
Platinum versus platinum-combination chemotherapy in platinum-sensitive recurrent ovarian cancer: a meta-analysis using individual patient data
.
Ann Oncol
2013
;
24
:
3028
34
.
66.
Parmar
MKB
,
Ledermann
JA
,
Colombo
N
,
du Bois
A
,
Delaloye
J-F
,
Kristensen
GB
, et al
.
Paclitaxel plus platinum-based chemotherapy versus conventional platinum-based chemotherapy in women with relapsed ovarian cancer: the ICON4/AGO-OVAR-2.2 trial
.
Lancet
2003
;
361
:
2099
106
.
67.
Di Maio
M
,
Chiodini
P
,
Georgoulias
V
,
Hatzidaki
D
,
Takeda
K
,
Wachters
FM
, et al
.
Meta-analysis of single-agent chemotherapy compared with combination chemotherapy as second-line treatment of advanced non-small-cell lung cancer
.
J Clin Oncol
2009
;
27
:
1836
43
.
68.
Wagner
AD
,
Grothe
W
,
Haerting
J
,
Kleber
G
,
Grothey
A
,
Fleig
WE
.
Chemotherapy in advanced gastric cancer: a systematic review and meta-analysis based on aggregate data
.
J Clin Oncol
2006
;
24
:
2903
9
.
69.
Van Cutsem
E
,
Moiseyenko
VM
,
Tjulandin
S
,
Majlis
A
,
Constenla
M
,
Boni
C
, et al
.
Phase III study of docetaxel and cisplatin plus fluorouracil compared with cisplatin and fluorouracil as first-line therapy for advanced gastric cancer: a report of the V325 Study Group
.
J Clin Oncol
2006
;
24
:
4991
7
.
70.
Xie
D-R
,
Liang
H-L
,
Wang
Y
,
Guo
S-S
,
Yang
Q
.
Meta-analysis on inoperable pancreatic cancer: a comparison between gemcitabine-based combination therapy and gemcitabine alone
.
World J Gastroenterol
2006
;
12
:
6973
81
.
71.
Moore
MJ
,
Goldstein
D
,
Hamm
J
,
Figer
A
,
Hecht
JR
,
Gallinger
S
, et al
.
Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group
.
J Clin Oncol
2007
;
25
:
1960
6
.
72.
Pusceddu
S
,
Ghidini
M
,
Torchio
M
,
Corti
F
,
Tomasello
G
,
Niger
M
, et al
.
Comparative effectiveness of gemcitabine plus nab-paclitaxel and FOLFIRINOX in the first-line setting of metastatic pancreatic cancer: a systematic review and meta-analysis
.
Cancers (Basel)
2019
;
11
:
484
.
73.
Centanni
M
,
Moes
DJAR
,
Trocóniz
IF
,
Ciccolini
J
,
van Hasselt
JGC
.
Clinical pharmacokinetics and pharmacodynamics of immune checkpoint inhibitors
.
Clin Pharmacokinet
2019
;
58
:
835
57
.
74.
Willsmore
ZN
,
Coumbe
BGT
,
Crescioli
S
,
Reci
S
,
Gupta
A
,
Harris
RJ
, et al
.
Combined anti-PD-1 and anti-CTLA-4 checkpoint blockade: treatment of melanoma and immune mechanisms of action
.
Eur J Immunol
2021
;
51
:
544
56
.
75.
Larkin
J
,
Chiarion-Sileni
V
,
Gonzalez
R
,
Grob
JJ
,
Cowey
CL
,
Lao
CD
, et al
.
Combined nivolumab and ipilimumab or monotherapy in untreated melanoma
.
N Engl J Med
2015
;
373
:
23
34
.
76.
Postow
MA
,
Chesney
J
,
Pavlick
AC
,
Robert
C
,
Grossmann
K
,
McDermott
D
, et al
.
Nivolumab and ipilimumab versus ipilimumab in untreated melanoma
.
N Engl J Med
2015
;
372
:
2006
17
.
77.
Schmid
P
,
Adams
S
,
Rugo
HS
,
Schneeweiss
A
,
Barrios
CH
,
Iwata
H
, et al
.
Atezolizumab and nab-paclitaxel in advanced triple-negative breast cancer
.
N Engl J Med
2018
;
379
:
2108
21
.
78.
Cortés
J
,
Cescon
DW
,
Rugo
HS
,
Im
S-A
,
Md Yusof
M
,
Gallardo
C
, et al
.
LBA16 KEYNOTE-355: final results from a randomized, double-blind phase III study of first-line pembrolizumab + chemotherapy vs placebo + chemotherapy for metastatic TNBC
.
Ann Oncol
2021
;
32
:
S1289
90
.
79.
Harrington
KJ
,
Burtness
B
,
Greil
R
,
Soulières
D
,
Tahara
M
,
de Castro
G
Jr
, et al
.
Pembrolizumab with or without chemotherapy in recurrent or metastatic head and neck squamous cell carcinoma: updated results of the phase III KEYNOTE-048 study
.
J Clin Oncol
2023
;
41
:
790
802
.
80.
Wainberg
ZA
,
Shitara
K
,
Van Cutsem
E
,
Wyrwicz
L
,
Lee
KW
,
Kudaba
I
, et al
.
Pembrolizumab with or without chemotherapy versus chemotherapy alone for patients with PD-L1–positive advanced gastric or gastroesophageal junction adenocarcinoma: update from the phase 3 KEYNOTE-062 trial
.
J Clin Oncol
2022
;
40
:
243
.
81.
Shitara
K
,
Van Cutsem
E
,
Bang
Y-J
,
Fuchs
C
,
Wyrwicz
L
,
Lee
K-W
, et al
.
Efficacy and safety of pembrolizumab or pembrolizumab plus chemotherapy vs chemotherapy alone for patients with first-line, advanced gastric cancer: the KEYNOTE-062 phase 3 randomized clinical trial
.
JAMA Oncol
2020
;
6
:
1571
80
.
82.
Janjigian
YY
,
Shitara
K
,
Moehler
M
,
Garrido
M
,
Salman
P
,
Shen
L
, et al
.
First-line nivolumab plus chemotherapy versus chemotherapy alone for advanced gastric, gastro-oesophageal junction, and oesophageal adenocarcinoma (CheckMate 649): a randomised, open-label, phase 3 trial
.
Lancet
2021
;
398
:
27
40
.
83.
Zou
K
,
Yang
S
,
Zheng
L
,
Yang
C
,
Xiong
B
.
Efficacy and safety of target combined chemotherapy in advanced gastric cancer: a meta-analysis and system review
.
BMC Cancer
2016
;
16
:
737
.
84.
Chen
P
,
Wang
L
,
Liu
B
,
Zhang
H-Z
,
Liu
H-C
,
Zou
Z
.
EGFR-targeted therapies combined with chemotherapy for treating advanced non-small-cell lung cancer: a meta-analysis
.
Eur J Clin Pharmacol
2011
;
67
:
235
43
.
85.
Ge
L
,
Tang
Y
,
Zhang
Q-N
,
Tian
J-H
,
Wang
X-H
,
Pieper
D
, et al
.
A network meta-analysis on the efficacy of targeted agents in combination with chemotherapy for treatment of advanced/metastatic triple-negative breast cancer
.
Oncotarget
2017
;
8
:
59539
51
.
86.
Housman
G
,
Byler
S
,
Heerboth
S
,
Lapinska
K
,
Longacre
M
,
Snyder
N
, et al
.
Drug resistance in cancer: an overview
.
Cancers (Basel)
2014
;
6
:
1769
92
.
87.
Longley
DB
,
Johnston
PG
.
Molecular mechanisms of drug resistance
.
J Pathol
2005
;
205
:
275
92
.
88.
Liu
N
.
Insecticide resistance in mosquitoes: impact, mechanisms, and research directions
.
Annu Rev Entomol
2015
;
60
:
537
59
.
89.
Sparks
TC
,
Dripps
JE
,
Watson
GB
,
Paroonagian
D
.
Resistance and cross-resistance to the spinosyns–a review and analysis
.
Pestic Biochem Physiol
2012
;
102
:
1
10
.
90.
Holderfield
M
,
Deuker
MM
,
McCormick
F
,
McMahon
M
.
Targeting RAF kinases for cancer therapy: BRAF-mutated melanoma and beyond
.
Nat Rev Cancer
2014
;
14
:
455
67
.
91.
Chong
CR
,
Jänne
PA
.
The quest to overcome resistance to EGFR-targeted therapies in cancer
.
Nat Med
2013
;
19
:
1389
400
.
92.
Khan
S
,
Uddin
MN
,
Rizwan
M
,
Khan
W
,
Farooq
M
,
Sattar Shah
A
, et al
.
Mechanism of insecticide resistance in insects/pests
.
Pol J Environ Stud
2020
;
29
:
2023
30
.
93.
Weetman
D
,
Mitchell
SN
,
Wilding
CS
,
Birks
DP
,
Yawson
AE
,
Essandoh
J
, et al
.
Contemporary evolution of resistance at the major insecticide target site gene Ace-1 by mutation and copy number variation in the malaria mosquito Anopheles gambiae
.
Mol Ecol
2015
;
24
:
2656
72
.
94.
Townsend
DM
,
Tew
KD
.
The role of glutathione-S-transferase in anti-cancer drug resistance
.
Oncogene
2003
;
22
:
7369
75
.
95.
Rochat
B
.
Role of cytochrome P450 activity in the fate of anticancer agents and in drug resistance: focus on tamoxifen, paclitaxel and imatinib metabolism
.
Clin Pharmacokinet
2005
;
44
:
349
66
.
96.
Rajagopal
R
,
Arora
N
,
Sivakumar
S
,
Rao
NGV
,
Nimbalkar
SA
,
Bhatnagar
RK
.
Resistance of Helicoverpa armigera to Cry1Ac toxin from Bacillus thuringiensis is due to improper processing of the protoxin
.
Biochem J
2009
;
419
:
309
16
.
97.
Vlogiannitis
S
,
Mavridis
K
,
Dermauw
W
,
Snoeck
S
,
Katsavou
E
,
Morou
E
, et al
.
Reduced proinsecticide activation by cytochrome P450 confers coumaphos resistance in the major bee parasite Varroa destructor
.
Proc Natl Acad Sci U S A
2021
;
118
:
e2020380118
.
98.
Gong
L
,
Kang
S
,
Zhou
J
,
Sun
D
,
Guo
L
,
Qin
J
, et al
.
Reduced expression of a novel midgut trypsin gene involved in protoxin activation correlates with Cry1Ac resistance in a laboratory-selected strain of plutella xylostella (L.)
.
Toxins (Basel)
2020
;
12
:
76
.
99.
Gottesman
MM
.
Mechanisms of cancer drug resistance
.
Annu Rev Med
2002
;
53
:
615
27
.
100.
Stewart
DJ
.
Mechanisms of resistance to cisplatin and carboplatin
.
Crit Rev Oncol Hematol
2007
;
63
:
12
31
.
101.
Dermauw
W
,
Wybouw
N
,
Rombauts
S
,
Menten
B
,
Vontas
J
,
Grbic
M
, et al
.
A link between host plant adaptation and pesticide resistance in the polyphagous spider mite Tetranychus urticae
.
Proc Natl Acad Sci U S A
2013
;
110
:
E113
22
.
102.
Gardiner
EMM
,
Plapp
FW
.
Insecticide uptake and decreased uptake resistance in the house fly (Diptera: Muscidae): a study with avermectin
.
J Econ Entomol
1997
;
90
:
261
6
.
103.
Heckel
DG
.
Learning the ABCs of Bt: ABC transporters and insect resistance to Bacillus thuringiensis provide clues to a crucial step in toxin mode of action
.
Pestic Biochem Physiol
2012
;
104
:
103
10
.
104.
Hernández-Martínez
P
,
Navarro-Cerrillo
G
,
Caccia
S
,
de Maagd
RA
,
Moar
WJ
,
Ferré
J
, et al
.
Constitutive activation of the midgut response to Bacillus thuringiensis in Bt-resistant Spodoptera exigua
.
PLoS One
2010
;
5
:
e12795
.
105.
Herrero
S
,
Gechev
T
,
Bakker
PL
,
Moar
WJ
,
de Maagd
RA
.
Bacillus thuringiensis Cry1Ca-resistant Spodoptera exigua lacks expression of one of four Aminopeptidase N genes
.
BMC Genomics
2005
;
6
:
96
.
106.
Bukowski
K
,
Kciuk
M
,
Kontek
R
.
Mechanisms of multidrug resistance in cancer chemotherapy
.
Int J Mol Sci
2020
;
21
:
3233
.
107.
Riganti
C
,
Contino
M
.
New strategies to overcome resistance to chemotherapy and immune system in cancer
.
Int J Mol Sci
2019
;
20
:
4783
.
108.
Hawkins
NJ
,
Bass
C
,
Dixon
A
,
Neve
P
.
The evolutionary origins of pesticide resistance
.
Biol Rev Camb Philos Soc
2019
;
94
:
135
55
.
109.
Clark
JM
,
Scott
JG
,
Campos
F
,
Bloomquist
JR
.
Resistance to avermectins: extent, mechanisms, and management implications
.
Annu Rev Entomol
1995
;
40
:
1
30
.
110.
Roush
R
,
Tabashnik
BE
.
Pesticide Resistance in Arthropods
.
Berlin (Germany)
:
Springer Science and Business Media
;
2012
.
111.
Labrie
M
,
Brugge
JS
,
Mills
GB
,
Zervantonakis
IK
.
Therapy resistance: opportunities created by adaptive responses to targeted therapies in cancer
.
Nat Rev Cancer
2022
;
22
:
323
39
.
112.
Vernieri
C
,
Prisciandaro
M
,
Milano
M
,
Cona
MS
,
Maggi
C
,
Brambilla
M
, et al
.
Single-agent gemcitabine vs. Carboplatin-gemcitabine in advanced breast cancer: a retrospective comparison of efficacy and safety profiles
.
Clin Breast Cancer
2019
;
19
:
e306
18
.
113.
Bai
X-F
,
Liu
J
,
Li
O
,
Zheng
P
,
Liu
Y
.
Antigenic drift as a mechanism for tumor evasion of destruction by cytolytic T lymphocytes
.
J Clin Invest
2003
;
111
:
1487
96
.
114.
de Vries
TJ
,
Fourkour
A
,
Wobbes
T
,
Verkroost
G
,
Ruiter
DJ
,
van Muijen
GN
.
Heterogeneous expression of immunotherapy candidate proteins gp100, MART-1, and tyrosinase in human melanoma cell lines and in human melanocytic lesions
.
Cancer Res
1997
;
57
:
3223
9
.
115.
Zaretsky
JM
,
Garcia-Diaz
A
,
Shin
DS
,
Escuin-Ordinas
H
,
Hugo
W
,
Hu-Lieskovan
S
, et al
.
Mutations associated with acquired resistance to PD-1 blockade in melanoma
.
N Engl J Med
2016
;
375
:
819
29
.
116.
Benci
JL
,
Xu
B
,
Qiu
Y
,
Wu
TJ
,
Dada
H
,
Twyman-Saint Victor
C
, et al
.
Tumor interferon signaling regulates a multigenic resistance program to immune checkpoint blockade
.
Cell
2016
;
167
:
1540
54.e12
.
117.
Saleh
R
,
Elkord
E
.
Treg-mediated acquired resistance to immune checkpoint inhibitors
.
Cancer Lett
2019
;
457
:
168
79
.
118.
Niederst
MJ
,
Engelman
JA
.
Bypass mechanisms of resistance to receptor tyrosine kinase inhibition in lung cancer
.
Sci Signal
2013
;
6
:
re6
.
119.
Giraudo
M
,
Califano
J
,
Hilliou
F
,
Tran
T
,
Taquet
N
,
Feyereisen
R
, et al
.
Effects of hormone agonists on Sf9 cells, proliferation and cell cycle arrest
.
PLoS One
2011
;
6
:
e25708
.
120.
Huang
J-F
,
Shui
K-J
,
Li
H-Y
,
Hu
M-Y
,
Zhong
G-H
.
Antiproliferative effect of azadirachtin A on Spodoptera litura Sl-1 cell line through cell cycle arrest and apoptosis induced by up-regulation of p53
.
Pestic Biochem Physiol
2011
;
99
:
16
24
.
121.
Sakka
MK
,
Riga
M
,
Ioannidis
P
,
Baliota
GV
,
Tselika
M
,
Jagadeesan
R
, et al
.
Transcriptomic analysis of s-methoprene resistance in the lesser grain borer, Rhyzopertha dominica, and evaluation of piperonyl butoxide as a resistance breaker
.
BMC Genomics
2021
;
22
:
65
.
122.
Mosallanejad
H
,
Smagghe
G
.
Biochemical mechanisms of methoxyfenozide resistance in the cotton leafworm Spodoptera littoralis
.
Pest Manag Sci
2009
;
65
:
732
6
.
123.
Rehan
A
,
Freed
S
.
Resistance selection, mechanism and stability of Spodoptera litura (Lepidoptera: Noctuidae) to methoxyfenozide
.
Pestic Biochem Physiol
2014
;
110
:
7
12
.
124.
Dawkar
VV
,
Barage
SH
,
Barbole
RS
,
Fatangare
A
,
Grimalt
S
,
Haldar
S
, et al
.
Azadirachtin-A from Azadirachta indica impacts multiple biological targets in cotton bollworm helicoverpa armigera
.
ACS Omega
2019
;
4
:
9531
41
.
125.
Xi
Y
,
Xu
P
.
Global colorectal cancer burden in 2020 and projections to 2040
.
Transl Oncol
2021
;
14
:
101174
.
126.
Xie
Y-H
,
Chen
Y-X
,
Fang
J-Y
.
Comprehensive review of targeted therapy for colorectal cancer
.
Signal Transduct Target Ther
2020
;
5
:
22
.
127.
Benson
AB
,
Venook
AP
,
Adam
M
,
Chang
G
,
Chen
Y-J
,
Ciombor
KK
, et al
.
Colon cancer, version 3.2024, NCCN clinical practice guidelines in oncology
.
J Natl Compr Canc Netw
2024
;
22
:
e240029
.
128.
Gatenby
RA
,
Brown
J
,
Vincent
T
.
Lessons from applied ecology: cancer control using an evolutionary double bind
.
Cancer Res
2009
;
69
:
7499
502
.
129.
Maley
CC
,
Reid
BJ
,
Forrest
S
.
Cancer prevention strategies that address the evolutionary dynamics of neoplastic cells: simulating benign cell boosters and selection for chemosensitivity
.
Cancer Epidemiol Biomarkers Prev
2004
;
13
:
1375
84
.
130.
Hamer
J
,
Warner
E
.
Lifestyle modifications for patients with breast cancer to improve prognosis and optimize overall health
.
CMAJ
2017
;
189
:
E268
74
.
131.
West
J
,
Adler
F
,
Gallaher
J
,
Strobl
M
,
Brady-Nicholls
R
,
Brown
JS
, et al
.
A survey of open questions in adaptive therapy: bridging mathematics and clinical translation
.
Elife
2023
;
12
:
e84263
.
132.
Lévi
F
,
Okyar
A
,
Dulong
S
,
Innominato
PF
,
Clairambault
J
.
Circadian timing in cancer treatments
.
Annu Rev Pharmacol Toxicol
2010
;
50
:
377
421
.
133.
Moloudi
K
,
Abrahamse
H
,
George
BP
.
Photodynamic therapy induced cell cycle arrest and cancer cell synchronization: review
.
Front Oncol
2023
;
13
:
1225694
.
134.
Zalatnai
A
.
Review: potential role of cell cycle synchronizing agents in combination treatment modalities of malignant tumors
.
In Vivo
2005
;
19
:
85
91
.
135.
Keir
ST
,
Friedman
HS
,
Reardon
DA
,
Bigner
DD
,
Gray
LA
.
Mibefradil, a novel therapy for glioblastoma multiforme: cell cycle synchronization and interlaced therapy in a murine model
.
J Neurooncol
2013
;
111
:
97
102
.
136.
Chari
RS
,
Tyler
DS
,
Anscher
MS
,
Russell
L
,
Clary
BM
,
Hathorn
J
, et al
.
Preoperative radiation and chemotherapy in the treatment of adenocarcinoma of the rectum
.
Ann Surg
1995
;
221
:
778
86
.
137.
Eisenhauer
EA
,
Therasse
P
,
Bogaerts
J
,
Schwartz
LH
,
Sargent
D
,
Ford
R
, et al
.
New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1)
.
Eur J Cancer
2009
;
45
:
228
47
.
138.
Cunningham
D
,
Chau
I
,
Stocken
DD
,
Valle
JW
,
Smith
D
,
Steward
W
, et al
.
Phase III randomized comparison of gemcitabine versus gemcitabine plus capecitabine in patients with advanced pancreatic cancer
.
J Clin Oncol
2009
;
27
:
5513
18
.
139.
Gatenby
RA
,
Silva
AS
,
Gillies
RJ
,
Frieden
BR
.
Adaptive therapy
.
Cancer Res
2009
;
69
:
4894
903
.
140.
Gatenby
RA
,
Brown
JS
.
Integrating evolutionary dynamics into cancer therapy
.
Nat Rev Clin Oncol
2020
;
17
:
675
86
.
141.
Seyedi
S
,
Teo
R
,
Foster
L
,
Saha
D
,
Mina
L
,
Northfelt
D
, et al
.
Testing adaptive therapy protocols using gemcitabine and capecitabine in a preclinical model of endocrine-resistant breast cancer
.
Cancers (Basel)
2024
;
16
:
257
.
142.
Thomas
DS
,
Cisneros
LH
,
Anderson
ARA
,
Maley
CC
.
In silico investigations of multi-drug adaptive therapy protocols
.
Cancers (Basel)
2022
;
14
:
2699
.
143.
El-Sayed
AM
,
Suckling
DM
,
Byers
JA
,
Jang
EB
,
Wearing
CH
.
Potential of “lure and kill” in long-term pest management and eradication of invasive species
.
J Econ Entomol
2009
;
102
:
815
35
.
144.
Are
C
,
Berman
RS
,
Wyld
L
,
Cummings
C
,
Lecoq
C
,
Audisio
RA
.
Global curriculum in surgical oncology
.
Ann Surg Oncol
2016
;
23
:
1782
95
.
145.
Rashid
OM
,
Nagahashi
M
,
Ramachandran
S
,
Graham
L
,
Yamada
A
,
Spiegel
S
, et al
.
Resection of the primary tumor improves survival in metastatic breast cancer by reducing overall tumor burden
.
Surgery
2013
;
153
:
771
8
.
146.
Hall
M
,
Savvatis
K
,
Nixon
K
,
Kyrgiou
M
,
Hariharan
K
,
Padwick
M
, et al
.
Maximal-effort cytoreductive surgery for ovarian cancer patients with a high tumor burden: variations in practice and impact on outcome
.
Ann Surg Oncol
2019
;
26
:
2943
51
.
147.
Coleman
RL
,
Spirtos
NM
,
Enserro
D
,
Herzog
TJ
,
Sabbatini
P
,
Armstrong
DK
, et al
.
Secondary surgical cytoreduction for recurrent ovarian cancer
.
N Engl J Med
2019
;
381
:
1929
39
.
148.
Khang
MK
,
Zhou
J
,
Huang
Y
,
Hakamivala
A
,
Tang
L
.
Preparation of a novel injectable in situ-gelling nanoparticle with applications in controlled protein release and cancer cell entrapment
.
RSC Adv
2018
;
8
:
34625
33
.
149.
Ko
C-Y
,
Wu
L
,
Nair
AM
,
Tsai
Y-T
,
Lin
VK
,
Tang
L
.
The use of chemokine-releasing tissue engineering scaffolds in a model of inflammatory response-mediated melanoma cancer metastasis
.
Biomaterials
2012
;
33
:
876
85
.
150.
de Groot
S
,
Pijl
H
,
van der Hoeven
JJM
,
Kroep
JR
.
Effects of short-term fasting on cancer treatment
.
J Exp Clin Cancer Res
2019
;
38
:
209
.
151.
Teleanu
RI
,
Chircov
C
,
Grumezescu
AM
,
Teleanu
DM
.
Tumor angiogenesis and anti-angiogenic strategies for cancer treatment
.
J Clin Med
2019
;
9
:
84
.
152.
Saxena
M
,
van der Burg
SH
,
Melief
CJM
,
Bhardwaj
N
.
Therapeutic cancer vaccines
.
Nat Rev Cancer
2021
;
21
:
360
78
.
153.
Wang
Y
,
Zhang
H
,
Liu
C
,
Wang
Z
,
Wu
W
,
Zhang
N
, et al
.
Immune checkpoint modulators in cancer immunotherapy: recent advances and emerging concepts
.
J Hematol Oncol
2022
;
15
:
111
.
154.
Waldman
AD
,
Fritz
JM
,
Lenardo
MJ
.
A guide to cancer immunotherapy: from T cell basic science to clinical practice
.
Nat Rev Immunol
2020
;
20
:
651
68
.
155.
Esfahani
K
,
Roudaia
L
,
Buhlaiga
N
,
Del Rincon
SV
,
Papneja
N
,
Miller
WH
Jr
.
A review of cancer immunotherapy: from the past, to the present, to the future
.
Curr Oncol
2020
;
27
(
Suppl 2
):
S87
97
.
156.
Martin
JH
,
Dimmitt
S
.
The rationale of dose-response curves in selecting cancer drug dosing
.
Br J Clin Pharmacol
2019
;
85
:
2198
204
.
157.
Brooks
EA
,
Galarza
S
,
Gencoglu
MF
,
Cornelison
RC
,
Munson
JM
,
Peyton
SR
.
Applicability of drug response metrics for cancer studies using biomaterials
.
Philos Trans R Soc Lond B Biol Sci
2019
;
374
:
20180226
.
158.
Tighiouart
M
.
Two-stage design for phase I–II cancer clinical trials using continuous dose combinations of cytotoxic agents
.
J R Stat Soc Ser C Appl Stat
2019
;
68
:
235
50
.
159.
Le Tourneau
C
,
Lee
JJ
,
Siu
LL
.
Dose escalation methods in phase I cancer clinical trials
.
J Natl Cancer Inst
2009
;
101
:
708
20
.
160.
Fraisse
J
,
Dinart
D
,
Tosi
D
,
Bellera
C
,
Mollevi
C
.
Optimal biological dose: a systematic review in cancer phase I clinical trials
.
BMC Cancer
2021
;
21
:
60
.
161.
Corbaux
P
,
El-Madani
M
,
Tod
M
,
Péron
J
,
Maillet
D
,
Lopez
J
, et al
.
Clinical efficacy of the optimal biological dose in early-phase trials of anti-cancer targeted therapies
.
Eur J Cancer
2019
;
120
:
40
6
.
162.
Gore
ME
,
Fryatt
I
,
Wiltshaw
E
,
Dawson
T
,
Robinson
BA
,
Calvert
AH
.
Cisplatin/carboplatin cross-resistance in ovarian cancer
.
Br J Cancer
1989
;
60
:
767
9
.
163.
Loria
R
,
Vici
P
,
Di Lisa
FS
,
Soddu
S
,
Maugeri-Saccà
M
,
Bon
G
.
Cross-resistance among sequential cancer therapeutics: an emerging issue
.
Front Oncol
2022
;
12
:
877380
.
164.
Bayat Mokhtari
R
,
Homayouni
TS
,
Baluch
N
,
Morgatskaya
E
,
Kumar
S
,
Das
B
, et al
.
Combination therapy in combating cancer
.
Oncotarget
2017
;
8
:
38022
43
.
165.
Leary
M
,
Heerboth
S
,
Lapinska
K
,
Sarkar
S
.
Sensitization of drug resistant cancer cells: a matter of combination therapy
.
Cancers (Basel)
2018
;
10
:
483
.
166.
Gatenby
RA
,
Brown
JS
.
The evolution and ecology of resistance in cancer therapy
.
Cold Spring Harb Perspect Med
2020
;
10
:
a040972
.
167.
Basanta
D
,
Gatenby
RA
,
Anderson
ARA
.
Exploiting evolution to treat drug resistance: combination therapy and the double bind
.
Mol Pharm
2012
;
9
:
914
21
.
168.
Vontas
J
,
Katsavou
E
,
Mavridis
K
.
Cytochrome P450-based metabolic insecticide resistance in Anopheles and Aedes mosquito vectors: muddying the waters
.
Pestic Biochem Physiol
2020
;
170
:
104666
.
169.
Buss
DS
,
Callaghan
A
.
Interaction of pesticides with p-glycoprotein and other ABC proteins: a survey of the possible importance to insecticide, herbicide and fungicide resistance
.
Pestic Biochem Physiol
2008
;
90
:
141
53
.
170.
Simmons
J
,
D’Souza
O
,
Rheault
M
,
Donly
C
.
Multidrug resistance protein gene expression in Trichoplusia ni caterpillars
.
Insect Mol Biol
2013
;
22
:
62
71
.
171.
Read
AF
,
Huijben
S
.
Evolutionary biology and the avoidance of antimicrobial resistance
.
Evol Appl
2009
;
2
:
40
51
.
172.
Foundation Cotton
.
Cotton insects and mites: characterization and management
.
Memphis (TN)
:
Cotton Foundation
;
1996
.
173.
Gu
S
,
Chakraborty
G
,
Champley
K
,
Alessio
AM
,
Claridge
J
,
Rockne
R
, et al
.
Applying a patient-specific bio-mathematical model of glioma growth to develop virtual [18F]-FMISO-PET images
.
Math Med Biol
2012
;
29
:
31
48
.
174.
West
J
,
You
L
,
Zhang
J
,
Gatenby
RA
,
Brown
JS
,
Newton
PK
, et al
.
Towards multidrug adaptive therapy
.
Cancer Res
2020
;
80
:
1578
89
.
175.
Basanta
D
,
Anderson
ARA
.
Homeostasis back and forth: an ecoevolutionary perspective of cancer
.
Cold Spring Harb Perspect Med
2017
;
7
:
a028332
.
176.
Fischer
A
,
Vázquez-García
I
,
Mustonen
V
.
The value of monitoring to control evolving populations
.
Proc Natl Acad Sci U S A
2015
;
112
:
1007
12
.
177.
Bürtin
F
,
Mullins
CS
,
Linnebacher
M
.
Mouse models of colorectal cancer: past, present and future perspectives
.
World J Gastroenterol
2020
;
26
:
1394
426
.
178.
Best
MG
,
Wesseling
P
,
Wurdinger
T
.
Tumor-educated platelets as a noninvasive biomarker source for cancer detection and progression monitoring
.
Cancer Res
2018
;
78
:
3407
12
.
179.
Roweth
HG
,
Battinelli
EM
.
Lessons to learn from tumor-educated platelets
.
Blood
2021
;
137
:
3174
80
.
180.
Torrano
V
,
Royo
F
,
Peinado
H
,
Loizaga-Iriarte
A
,
Unda
M
,
Falcón-Perez
JM
, et al
.
Vesicle-MaNiA: extracellular vesicles in liquid biopsy and cancer
.
Curr Opin Pharmacol
2016
;
29
:
47
53
.
181.
Siravegna
G
,
Marsoni
S
,
Siena
S
,
Bardelli
A
.
Integrating liquid biopsies into the management of cancer
.
Nat Rev Clin Oncol
2017
;
14
:
531
48
.
182.
Armakolas
A
,
Kotsari
M
,
Koskinas
J
.
Liquid biopsies, novel approaches and future directions
.
Cancers (Basel)
2023
;
15
:
1579
.
183.
Keller
L
,
Belloum
Y
,
Wikman
H
,
Pantel
K
.
Clinical relevance of blood-based ctDNA analysis: mutation detection and beyond
.
Br J Cancer
2021
;
124
:
345
58
.
184.
Abbosh
C
,
Birkbak
NJ
,
Wilson
GA
,
Jamal-Hanjani
M
,
Constantin
T
,
Salari
R
, et al
.
Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution
.
Nature
2017
;
545
:
446
51
.
185.
Cao
H
,
Liu
X
,
Chen
Y
,
Yang
P
,
Huang
T
,
Song
L
, et al
.
Circulating tumor DNA is capable of monitoring the therapeutic response and resistance in advanced colorectal cancer patients undergoing combined target and chemotherapy
.
Front Oncol
2020
;
10
:
466
.
186.
Murtaza
M
,
Dawson
S-J
,
Pogrebniak
K
,
Rueda
OM
,
Provenzano
E
,
Grant
J
, et al
.
Multifocal clonal evolution characterized using circulating tumour DNA in a case of metastatic breast cancer
.
Nat Commun
2015
;
6
:
8760
.
187.
Pessoa
LS
,
Heringer
M
,
Ferrer
VP
.
ctDNA as a cancer biomarker: a broad overview
.
Crit Rev Oncol Hematol
2020
;
155
:
103109
.
188.
Chen
K-Z
,
Lou
F
,
Yang
F
,
Zhang
J-B
,
Ye
H
,
Chen
W
, et al
.
Circulating tumor DNA detection in early-stage non-small cell lung cancer patients by targeted sequencing
.
Sci Rep
2016
;
6
:
31985
.
189.
Abbosh
C
,
Birkbak
NJ
,
Wilson
GA
,
Jamal-Hanjani
M
,
Constantin
T
,
Salari
R
, et al
.
Corrigendum: phylogenetic ctDNA analysis depicts early-stage lung cancer evolution
.
Nature
2018
;
554
:
264
.
190.
Herberts
C
,
Annala
M
,
Sipola
J
,
Ng
SWS
,
Chen
XE
,
Nurminen
A
, et al
.
Deep whole-genome ctDNA chronology of treatment-resistant prostate cancer
.
Nature
2022
;
608
:
199
208
.
191.
Luo
H
,
Zhao
Q
,
Wei
W
,
Zheng
L
,
Yi
S
,
Li
G
, et al
.
Circulating tumor DNA methylation profiles enable early diagnosis, prognosis prediction, and screening for colorectal cancer
.
Sci Transl Med
2020
;
12
:
eaax7533
.
192.
Chen
X
,
Gole
J
,
Gore
A
,
He
Q
,
Lu
M
,
Min
J
, et al
.
Non-invasive early detection of cancer four years before conventional diagnosis using a blood test
.
Nat Commun
2020
;
11
:
3475
.
193.
Budhraja
KK
,
McDonald
BR
,
Stephens
MD
,
Contente-Cuomo
T
,
Markus
H
,
Farooq
M
, et al
.
Genome-wide analysis of aberrant position and sequence of plasma DNA fragment ends in patients with cancer
.
Sci Transl Med
2023
;
15
:
eabm6863
.
194.
Lone
SN
,
Nisar
S
,
Masoodi
T
,
Singh
M
,
Rizwan
A
,
Hashem
S
, et al
.
Liquid biopsy: a step closer to transform diagnosis, prognosis and future of cancer treatments
.
Mol Cancer
2022
;
21
:
79
.
195.
Ossandon
MR
,
Agrawal
L
,
Bernhard
EJ
,
Conley
BA
,
Dey
SM
,
Divi
RL
, et al
.
Circulating tumor DNA assays in clinical cancer research
.
J Natl Cancer Inst
2018
;
110
:
929
34
.
196.
Marusyk
A
,
Polyak
K
.
Tumor heterogeneity: causes and consequences
.
Biochim Biophys Acta
2010
;
1805
:
105
17
.
197.
Hughes
D
,
Andersson
DI
.
Evolutionary consequences of drug resistance: shared principles across diverse targets and organisms
.
Nat Rev Genet
2015
;
16
:
459
71
.
198.
Kuosmanen
T
,
Cairns
J
,
Noble
R
,
Beerenwinkel
N
,
Mononen
T
,
Mustonen
V
.
Drug-induced resistance evolution necessitates less aggressive treatment
.
PLoS Comput Biol
2021
;
17
:
e1009418
.
199.
Huijben
S
,
Chan
BHK
,
Nelson
WA
,
Read
AF
.
The impact of within-host ecology on the fitness of a drug-resistant parasite
.
Evol Med Public Health
2018
;
2018
:
127
37
.
200.
Read
AF
,
Day
T
,
Huijben
S
.
The evolution of drug resistance and the curious orthodoxy of aggressive chemotherapy
.
Proc Natl Acad Sci U S A
2011
;
108
(
Suppl 2
):
10871
7
.
This open access article is distributed under the Creative Commons Attribution 4.0 International (CC BY 4.0) license.