- Review
- Open access
- Published:
Decoding the mechanisms behind second primary cancers
Journal of Translational Medicine volume 23, Article number: 115 (2025)
Abstract
Second Primary Cancers (SPCs) are defined as cancers that develop either simultaneously or metachronously in the same individual who has been diagnosed with and survived one primary cancer. SPCs exhibit a high incidence rate and represent the primary cause of mortality among survivors of first primary cancers. There is growing concern about the dangers of SPCs. This review summarizes recent studies on the mechanisms of SPCs, including the roles of genomic changes after first primary cancer (FPC) treatments, stromal cell phenotypic and metabolic changes, hormone levels and receptor expression, immunosuppression, aberrant gene methylation, EGFR signaling, and cell-free DNA in SPC development. This comprehensive analysis contributes to elucidating current research trends in SPC mechanisms and enhances our understanding of the underlying pathophysiology. Furthermore, potential applications of intratumoral microbes, single-cell multi-omics, and metabolomics in investigating SPC mechanisms are also discussed, providing new ideas for follow-up studies.
Introduction
Second Primary Cancers (SPCs) are defined as cancers that develop either simultaneously or metachronously in the same individual who has been diagnosed with and survived one primary cancer [1]. The significance of SPCs stems from their high incidence and substantial impact on first primary cancer (FPC) survivors. The incidence of SPCs varies widely, ranging from 2.4% to 19.0% [2,3,4]. A previous meta-analysis, encompassing a pooled cohort of 9,617,643 cancer patients, of whom 677,430 developed SPCs, examined the absolute numbers of patients with various types of FPCs who subsequently developed SPCs. The study revealed that among 1,825,198 breast cancer patients, 1,181,621 prostate cancer patients, and 711,504 skin cancer patients, 117,178, 111,150, and 76,517 respectively developed SPCs, ranking these three cancer types as the most prevalent in terms of SPC occurrence. Upon analyzing the incidence of SPCs among patients with various FPCs, the study revealed that patients with primary ureteral cancer exhibited the highest SPC incidence of 27.0%, followed by those with primary penis cancer (17.6%) and primary bladder cancer (15.8%) [5]. FPC survivors face an SPC risk 1.27 to 2.6 times higher than that of the general population [6, 7], with a 10–14% increased likelihood of developing SPCs [8, 9]. SPCs significantly impact FPC mortality, increasing the risk of death for FPC survivors by 33% in women and 45% in men.SPCs represent a leading cause of mortality among FPC survivors [10].
While SPC pose a significant threat to patients who have survived FPC, the precise mechanisms underlying SPC tumorigenesis remain a subject of debate. The molecular pathways involved in the development of general cancers encompass genetic mutations, aberrant signaling pathways, epigenetic alterations, tumor microenvironment as well as immune factors [11,12,13,14,15,16,17,18], all contributing synergistically to tumor initiation and progression.
While FPCs and SPCs share certain similarities in their pathogenesis, there are also notable differences between them. The key differences in their mechanisms can be summarized as follows:
Treatment-Related Factors: The emergence of SPCs may be intricately linked to the adverse effects of cancer therapies, particularly chemotherapy and radiotherapy. These treatments, while targeting and eliminating FPC, can inadvertently damage normal cells, thereby elevating the risk of SPC development. For instance, peroxides generated during FPC radiotherapy have been implicated in SPC induction [19]. Radiation-cell interactions lead to the production of free radicals that convert to peroxides, which then infiltrate unaffected cells [20, 21], inducing chromosomal damage and consequently radiation-induced bystander genetic damage, potentially contributing to SPCs.
Immune System Status: While the onset of FPCs may be associated with immune dysfunction, the development of SPCs can be further influenced by the immunomodulatory effects of cancer treatments. Immune cells and cytokines play pivotal roles in SPC development [20]. During FPC radiotherapy, lymphocytes and macrophages stimulated by radiation [22] amplify the production of inflammatory cytokines (IL-1, IL-2, IL-6, IL-8, TNFα, TGFβ) in unaffected cells. These cytokines promote SPC development post-radiotherapy by stimulating cell growth/proliferation, inhibiting cancer cell apoptosis, recruiting type 1 macrophages to augment inflammatory responses, and modulating immune responses [23,24,25,26,27,28].
Further Implications of Genetic Factors: Beyond their role in FPC development, genetic factors may continue to influence the emergence of SPCs. Hereditary tumor susceptibility genes in patients with hereditary tumor syndromes have been implicated in the development of SPCs or multiple primary cancers[29]. For example, germline p53 mutations in Li-Fraumeni syndrome patients increase the risks for various cancers, including breast cancer, osteosarcoma, soft tissue sarcoma, brain tumors, adrenocortical carcinoma, and leukemia [30,31,32,33,34].
Although SPCs significantly impact FPC survivor survival and quality of life, the mechanisms underlying SPC occurrence remain controversial. Some studies suggest that immune cells and cytokines play key roles in SPC development, while others propose that hereditary tumor susceptibility genes underlie SPC formation. To date, no comprehensive review has systematically examined SPC mechanisms. Therefore, this review summarizes recent studies on SPC mechanisms and discusses the potential applications of intratumoral microbiology, single-cell multi-omics, and metabolomics in exploring SPC mechanisms, thereby providing insights for future research.
Risk factors of second primary carcinogenesis
Summarizing the risk factors associated with second primary cancers can help elucidate the potential relationships between multiple primary cancers and their common pathogenic factors. It is beneficial to further explore the molecular mechanism and biological pathway underlying second primary cancer development. We have identified and categorized the SPC risk factors including: tobacco and alcohol, obesity and diet, virus infection, genetic predisposition and pathways, and treatment-related factors.
Tobacco and alcohol
In a study of 25,000 subsequent cancers occurring in individuals with primary tumors at sites associated with tobacco and alcohol use (e.g., oral cavity, pharynx, esophagus, larynx, lung, and bronchus), more than 11,000 recurred at sites linked to tobacco and alcohol consumption. This finding underscores the enduring relationship between these substances and the development of second primary cancers [35].
Tobacco
Tobacco exposure induces the expression of urokinase-type plasminogen activator (uPA), leading to plasmin-dependent degradation of the extracellular matrix (ECM) and enhanced cell migration. This mechanism may contribute to the development of multiple primary tumors in patients with head and neck cancer [36]. Continued smoking following successful treatment of head and neck cancer significantly elevates the risk of developing second primary cancers (SPCs) [37,38,39]. This increased risk may be attributed to tobacco-induced activation of the epidermal growth factor receptor (EGFR) pathway [40]. EGFR stimulation activates ERK1/2, increasing uPA expression [41, 42]. uPA cleaves plasminogen into ECM-degrading plasmin [43, 44], promoting tumor cell invasion and metastasis [36, 45, 46]. This mechanism may contribute to SPC development with a common clonal origin in smoking head and neck cancer patients [47,48,49]. Since uPA-uPAR binding stimulates EGFR and downstream ERK signaling [50] tobacco may activate an EGFR-ERK-uPA feedback loop, amplifying its carcinogenic effects.
Alcohol
The interaction between genetic factors, environmental alcohol consumption, and the ALDH2-2 allele increases the risk of multiple Lugol-voiding lesions (LVLs), which are characterized by dysplastic or hyperkeratinized unstained epithelium [51]. This interaction subsequently drives the development of second primary esophageal squamous cell carcinoma (ESCC) in patients with head and neck squamous cell carcinoma (HNSCC). The aldehyde dehydrogenase 2 (ALDH2) gene allele (ALDH2-2) is strongly associated with the occurrence of multiple LVLs [52, 53]. Chronic drinkers with the ALDH2-2 allele, who lack compensatory mechanisms for acetaldehyde elimination, exhibit elevated levels of LVLs [54]. As high acetaldehyde exposure from exhalation may contribute to the development of multiple LVLs [52].
HNSCC patients frequently develop second primary ESCCs [54,55,56], and multiple LVLs associate with high head and neck/esophageal multiple cancer risk [57, 58]and predict ESCC onset. Consequently, the presence of multiple LVLs may facilitate the development of secondary ESCCs in patients with HNSCC [52, 59,60,61]. In conclusion, alcohol consumption induces the formation of multiple LVLs in patients carrying the ALDH2-2 allele through complex genetic and environmental interactions. This process subsequently increases the susceptibility to second primary ESCCs in HNSCC patients [53].
Obesity and diet
Obesity is a well-established risk factor for multiple cancers, such as endometrial, colon, esophageal, kidney, pancreatic, and postmenopausal breast cancers, and is implicated in SPC development [62]. Moreover, factors including obesity, physical inactivity, and reproductive characteristics play a crucial role in the development of hormone-responsive tumors, particularly malignancies of the breast, uterus, ovaries, and prostate, as well as colorectal cancer [35]. Although the direct association between diet and SPCs remains incompletely elucidated, dietary factors, in conjunction with alcohol and tobacco consumption, exacerbate the risk of SPCs in patients with oral and pharyngeal cancers [63].
Virus infection
Viral infections, particularly Human papillomavirus (HPV), human immunodeficiency virus (HIV), human herpesvirus 8, Epstein-Barr virus (EBV), hepatitis B and C viruses, and Helicobacter pylori, play crucial roles in the development of SPCs [35, 64]. For instance, patients with one HPV-related cancer exhibit an elevated risk of developing another HPV-related SPC. Additionally, HPV infection is implicated in the concurrent occurrence of cervical and anogenital tract cancers, as well as oral cancers [62].
Genetic predisposition and pathways
Patients with hereditary cancer susceptibility syndromes carrying germline mutations exhibit an elevated risk of developing primary tumors at multiple sites, indicating a potential association between these germline mutations and SPC. For example, hereditary cancer predisposition syndromes, such as Lynch syndrome (formerly known as hereditary non-polyposis colorectal cancer or HNPCC), are associated with an elevated risk of SPCs in multiple organs, including the uterus, ovaries, bile ducts, small intestine, and renal pelvis. These syndromes are frequently associated with mutations in DNA mismatch repair genes, particularly MSH2 and MLH1 [65].
Furthermore, single nucleotide polymorphisms (SNPs) have also been implicated in an increased risk of developing SPC. Comparing individuals with multiple primary cancers versus cancer-free controls identified 22 potentially associated mutations. Of these, 10 mutations remained statistically significant when comparing patients with multiple cancers to those with a single cancer [66]. For instance, the rs7872034 (SMC2 missense variant) and rs143745791 (NCBP1 missense variant) showed stronger associations with the combination of primary breast cancer and any additional cancer compared to cancer-free individuals. These variants also demonstrated significant associations with the occurrence of breast cancer along with additional cancers, as opposed to breast cancer alone. The burden of oncogenic SNPs increased with co-morbid thyroid and breast cancers compared to just breast cancer [67]. Considering the apoptotic role of FAS/FASLG [68, 69] and the associations between their polymorphisms and cancer risk [70,71,72,73], Lei et al. [74] hypothesized that FAS/FASLG polymorphisms might influence the risk of developing SPCs. Indeed, head and neck squamous cell carcinoma (HNSCC) patients with FAS-670 AG/GG or FASLG-844 CT/TT variant genotypes had significantly higher SPC risk, increasing with more combined risk genotypes. Thus, FAS/FASLG polymorphisms could potentially serve as biomarkers for assessing SPC risk in HNSCC patients. Additionally, studies have demonstrated that p21 polymorphisms in HNSCC patients are also associated with an increased risk of developing SPCs [75,76,77].
Moreover, specific signaling pathways have been implicated in the development of SPCs. For example, in patients with concurrent malignant melanoma and renal cell carcinoma, genetic variations in the PI3K/mTOR signaling pathway have been identified as a potentially significant common risk factor [78,79,80].
For easy reference, we've summarized key germline mutations, single nucleotide polymorphisms (SNPs), and signaling pathways related to SPCs in Tables 1, 2, 3.
Treatment
Treatment for FPC, particularly radiation therapy and certain hormone therapies, can also contribute to SPC development. For instance, radiation therapy for breast cancer increases the risk of lung, esophageal, and sarcoma development. Similarly, pelvic radiation therapy for cervical and uterine cancers has been associated with an elevated risk of acute leukemia [35]. In patients with retinoblastoma, mutations in the RB1 gene enhance radiosensitivity, thereby predisposing these individuals to radiation-induced malignancies such as osteosarcomas, soft-tissue sarcomas, and melanomas [81]. Additionally, Hodgkin's lymphoma survivors undergoing radiotherapy face a substantial risk of developing breast, lung, and other cancers [82]. Certain hormone therapies, such as tamoxifen used in breast cancer treatment, have been associated with an increased risk of endometrial cancer [35].
Mechanisms of second primary carcinogenesis
Genomic changes
DNA damage
Radiotherapy-induced DNA single-strand breaks (SSBs) can initiate senescence and serve as a mechanism for SPC initiation. Senescent cells may escape cell-cycle arrest, giving rise to mutant and invasive daughter cells with cancerous characteristics [83]. Standard radiotherapy protocols targeting FPCs [84] produce off-site doses resulting in SSB accumulation in normal fibroblasts at the planning target volume (PTV) edge [85]. Reduced poly(ADP-ribosyl)ation (PARylation) capacity impairs SSB repair [86, 87]. In fibroblasts, the accumulation of unrepaired SSBs does not cause death but upregulates senescence-associated p16, inducing early senescence [88,89,90,91].
Unlike persistent double-strand breaks (DSBs) causing permanent tumor-suppressor cell cycle arrest [92,93,94,95,96,97], radiotherapy-induced persistent SSBs allow mutation and senescence escape [90, 98]. Cells with persistent SSBs initially senesce but some re-enter the cell cycle, producing daughter cells with precancerous but not fully tumourigenic features [99, 100]. These daughter cells express precancerous transformation biomarkers, such as post-senescence neoplastic emergence (PSNE) genes and epithelial-mesenchymal transition (EMT) markers, and can form small carcinomas in mice, demonstrating oncogenic potential [99, 101,102,103].
Consequently, SSB-induced cellular senescence may play a role in initiating SPCs.
Telomere damage
Patients with shortened or damaged telomeres following chemotherapy or radiotherapy may be at an elevated risk of developing SPC [104]. First primary cancer (FPC) therapy results in significant telomere shortening, reduced telomerase activity, and decreased expression of human telomerase reverse transcriptase (hTERT) and telomere-binding proteins (TPP1, POT1)) [105]. Samples exposed to chemotherapy exhibit shortened telomeres and reduced telomerase activity compared to untreated samples [106].Patients undergoing chemotherapy demonstrate shorter telomeres compared to healthy controls [107]. Ionizing radiation has also been shown to impair telomere integrity in vivo [108, 109].
Telomere loss from radiotherapy causes chromosomal instability, mainly through the breakage-fusion-bridge (B/F/B) cycle (Fig. 1). Telomere-free fused sister chromatids form bridges during replication that break when pulled to daughter cells. One daughter then has an inverted duplication chromosome end lacking a telomere, while the other have a deletion at the end. Both replicate aberrantly, continuing the B/F/B cycle. Besides end amplifications, telomere loss can cause arm amplifications and transfer anomalies to other chromosomes, elevating instability [110,111,112].
Telomere loss-induced chromosome instability related to the breakage-fusion-bridge (B/F/B) cycle after radiotherapy and chemotherapy. First primary cancer radiotherapy and chemotherapy cause telomere breakage. Replicating telomere-free fused sister chromatids form bridges that break when pulled apart, causing one daughter cell to have an inverted duplication end lacking a telomere, while the other to have a deletion at the end. With continued telomere loss, B/F/B cycles accumulate inverted chromosome end repeats and progressive end deletions. Pink, green, and red arrows indicate subtelomeric sequence direction
Telomere damage-induced genomic instability may enable cancer evolution by accelerating genetic changes [113, 114].Firstly, B/F/B cycles create daughter cells with inverted repeat chromosome ends associated with gene amplification [115] and cancer [116,117,118].Second, short telomeres associate with increased head and neck, bladder, lung, and renal cancers [119]. Familial thyroid cancer patients show more telomere damage than spontaneous cases, potentially underlying cancer-enabling genomic instability [120, 121]. Thus, FPC therapy-induced telomere damage may promote second carcinogenesis through genomic instability.
Phenotypic and metabolic changes in stromal cells
Phenotypic and metabolic alterations in stromal cells contribute significantly to the development of SPC [122] (Fig. 2). Chemotherapy induces oxidative stress in stromal cells adjacent to cancer cells, activating stress-related signaling pathways (e.g., HIF, NF-κB, SMAD, STAT3, JNK/AP1) [123]. This activation promotes various cellular processes, including glycolysis, mitochondrial dysfunction, autophagy, senescence, and the release of inflammatory cytokines. This stimulates cancer-associated fibroblast (CAF) differentiation, which facilitates tumor occurrence, progression, and metastasis [123,124,125]. Glycolytic/autophagic pathway upregulation in stromal fibroblasts increases energy-rich metabolite (e.g. lactate, ketones, glutamine) production, fueling cancer cell metabolic efficiency and promoting apoptosis resistance [126, 127]. Additionally, damaged fibroblasts promote the secretion of cytokines, such as IL-6, which aid in cancer cell survival and metastasis. Thus, metabolite-enriched microenvironments may enable malignant transformation of post-chemotherapy partially transformed epithelial cells, spurring new tumors [128, 129].
Role of chemotherapy-induced cancer-associated fibroblast (CAF) in transforming partially transformed epithelial cells into second primary cancers (SPCs). In CAF-contacting partially transformed epithelial cells, pathways such as Shh, Wnt, and STAT3 involved in stem/progenitor cell renewal, metabolism, metastasis, and drug resistance may be activated. CAFs also produce metabolites like lactate, ketones, glutamine, fatty acids, and cysteine that fuel epithelial cell SPC transformation
Chemotherapy also activates pathways related to stem/progenitor cell renewal, metabolism, metastasis, and chemoresistance (including Shh, Wnt, TGF-β, and STAT3) in breast cancer cells that are in contact with stromal cells [122]. Shh pathway activation associates with cancer development [130]. STAT3/Wnt pathways enhance oxidative phosphorylation, further promoting cancer cell growth [131, 132]. CAFs may stimulate stemness in impaired precancerous epithelial cells via Shh/Wnt/STAT3 pathway activation, triggering new oncogenic process [122].
Hormones
Serum insulin-like growth factor 1 (IGF-1)
Elevated serum IGF-1 levels are associated with an increased risk of second primary epithelial cancers (e.g., breast, lung, colon) in patients with head and neck cancer. Higher IGF-1 levels significantly increase SPC development risk [133]. In a breast cancer trial, lower IGF-1 levels were associated with fewer second primary cancers in premenopausal women [134]. The proposed mechanisms by which IGF-1 contributes to second primary cancer development include: (1) enhancing cell proliferation, apoptosis resistance, and clonal expansion of genetically damaged populations [135,136,137,138]; (2) regulating anti-apoptotic Bcl-xL/pro-apoptotic Bax ratios, affecting cell survival [139]; (3) increasing vascular endothelial growth factor (VEGF) (production as an angiogenesis regulator [140, 141]; (4) paracrine/endocrine promotion of second breast cancer growth by stromal cell-expressed IGF-1 [142]. Younger Hodgkin's lymphoma females have higher second breast cancer risk than older women [143, 144]. IGF-1 and growth hormone (GH) mediate pubertal mammary gland growth via estrogen [145,146,147]. Thus, Shanmugalingam et al. [142]hypothesized that elevated IGF-1/estrogen during puberty and stromal expressed IGF-1 may promote second breast cancer development, progression and metastasis by inhibiting apoptosis and inducing survival [148,149,150]. (5) Lung stromal cell expressed IGF-1 may act similarly through paracrine effects on bronchial epithelium in second lung cancers[142]. (6) Radiation-generated colon transformed cells may survive via increased IGF-1 bioactivity, enabling subsequent carcinogenesis through IGF-1R activation of proliferative, anti-apoptotic pathways, as in normal colon tissues, IGF-1 has a high affinity for IGF-1R and activates specific insulin receptor substrates [151].
Estrogen
High estrogen levels or estrogen receptor (ER) overexpression may contribute to concurrent thyroid and breast cancer [67]. Recent decades show increased breast cancer incidence associated with heightened exogenous/endogenous estrogens [152,153,154]. Estrogen binds ERα, regulating proliferation genes through genomic and non-genomic actions. ERα also enhances cancer cell survival/adaptation by responding to stresses and conditions [155]. Estrogen/receptor roles in thyroid cancer pathogenesis/progression include: ER overexpression in thyroid cancer [156, 157]; estrogen stimulation of benign/malignant thyroid cell growth in vitro [158,159,160]; estrogen-induced thyroid tumor cells’ growth via mitogen-activated protein kinase (MAPK) activation [161, 162]; and involvement in the thyroid tumor microenvironment [163]. Thus, elevated estrogen/ER may underlie thyroid-breast cancer co-morbidity.
Indeed, estrogen/progesterone receptors were significantly higher in breast cancer specimens from women with co-occurring differentiated thyroid cancer versus controls with only breast cancer, regardless of cancer order [164,165,166]. ERa in the breast and thyroid can be activated by the MAPK pathway, promoting carcinogenesis [160, 167, 168]. Estrogen also binds GPR30 on the endoplasmic reticulum, activating MAPK pathway and thyroid/breast cancers [169, 170]. Overexpression of COMP may enable thyroid/breast cancer tumorigenesis/progression through estrogen signaling [171].
Immune suppression
Immunosuppression may increase second primary cancer risk. In addition to chemoradiotherapy effects, the association between secondary non-Hodgkin lymphoma (NHL) (a known immunosuppression-linked cancer [172, 173]) and melanoma/skin squamous cell carcinoma treated by surgery suggests immunosuppression promotes SPCs [174]. The first primary tumor may further impair immunity via chronic inflammation and defense mechanism suppression [175], enabling malignant cell immune evasion [176, 177]. Such immunosuppression may contribute to excessive SPC risk [178]. First primary cancer/treatment immunosuppression may also cause endogenous viral reactivation elsewhere, spurring second NHLs since Epstein-Barr virus (EBV) plays a key NHL infectious role [173] and post-transplant EBV reactivation drives post-transplant carcinogenesis [179]. Thus, immunosuppression may promote SPCs by facilitating the escape of malignant cells from immune surveillance and enabling viral reactivation.
Other mechanisms
Abnormal gene methylation
Abnormal gene methylation contributes to SPC development. For example, tissue inhibitor of metalloproteinase 3 (TIMP3) hypermethylation and transcriptional repression impedes its matrix metalloproteinase (MMP) inhibition. Since the extracellular matrix (ECM) enables malignant spread [180], this may promote invasion, tumorigenesis, metastasis and angiogenesis [181]. Thus, TIMP3 inhibition by hypermethylation/loss of MMP regulatory ability may promote SPCs [182].
Indeed, TIMP3 hypermethylation associated significantly with local recurrence-free survival in head and neck squamous cell carcinoma (HNSCC) [183]. Increased MMP9 expression in HNSCC histologically negative surgical margins also associated with SPC development [184], supporting hypermethylated TIMP3 enables SPC development through MMP disinhibition.
Hypermethylation of promoter regions often silences genes, causing loss of function. CCNA1 promoter hypermethylation inhibits apoptosis and cell cycle arrest by reducing CCNA1 expression. This promotes precancerous cell proliferative dominance and expansion of carcinoma-prone progenitor cells, enabling carcinogenesis [182].
EGFR signaling
Breast and lung cancers can co-occur as second primaries, and EGFR mutations may contribute to their concurrence [185]. Second primary lung cancers tend to follow first primary breast cancers [185, 186]. Zeng et al. observed a two-fold higher EGFR mutation rate in breast cancer patients with secondary lung cancer versus other cancer patients with secondary lung cancer, suggesting EGFR signaling may play a key role in concurrent breast-lung cancer[185]. Proposed EGFR signaling mechanisms in concurrent breast-lung cancer include:
-
(1) In solid tumors including breast cancer, EGFR pathway dysregulation via receptor/ligand overexpression, phosphatase deficiency, and altered dimerization causes carcinogenesis [187,188,189].
-
(2) EGFR can activate estrogen receptor (ER) signaling, which is important in primary lung cancer after breast cancer [185, 190, 191].
-
(3) EGFR is a growth factor receptor that activates intracellular cancer-associated signaling pathways [192, 193].
-
(4) EGFR overexpression associates with apoptosis and tumor angiogenesis [194, 195].
In summary, EGFR mutations may provide insights into the common cause of concurrent breast-lung cancers.
Circulating cell-free DNA
Chen et al.'s in vitro mouse tumor growth experiments after injecting normal cells transformed by free DNA suggest oncogenic circulating cell-free DNA (cf-DNA) may act like an intrinsic oncolytic virus [196]. Released by dividing cancer cells into body fluids, cf-DNA may transfect or transform adjacent/distant normal cells. Transformed cells may then proliferate into second primary cancers (Fig. 3).
First primary cancer (FPC) cell-free DNA induction of second primary cancers (SPCs) via normal cell transfection/transformation. FPC cells release circulating cell-free DNA via apoptosis, necrosis, autophagy, and division. This cell-free DNA transfects or transforms distant normal cells, inducing SPC formation
Cf-DNA primarily leaks from apoptotic/necrotic cells into blood [197,198,199]. However, contrary to expectations of chemotherapy-induced apoptosis increasing cf-DNA, it significantly decreased cf-DNA [200]. Thus, Chen et al. speculated proliferating cancer cells may also release DNA during proliferation [196]. Thierry summarized three cf-DNA sources in cancer patients based on increased blood cf-DNA levels correlating with tumor cell numbers: healthy cells, malignant cells, tumor microenvironmental cells. Proliferation, apoptosis, necrosis, or secretion of cancer cell release cancer cell cf-DNA into circulation through structures like exosomes, microparticles, apoptotic bodies, nucleosomes, proteolipidonucleic acid complexes, DNA traps, or serum protein/cell membrane links [201].
Indeed, injected tumor DNA transformed mice [202], leading to the "genomic arrest" hypothesis where tumor-released cf-DNA transfects distant healthy cells, enabling metastasis [201, 203]. P. Anker et al. found after adding plasma from colorectal cancer mutant KRAS carriers (containing cf-DNA) to the culture medium, NIH/3T3 cells became carriers of both tumor and human KRAS mutations [204]. Patients who underwent resection of primary colorectal tumors with detectable tumor cf-DNA in plasma that can transform NIH-2T29 cells subsequently developed liver and lung cancers [205]. Thus, Chen hypothesized proliferating cancer cell-released cf-DNA may transfect or transform normal cells, explaining second primary cancer onset [196].
Prospects
The etiology and mechanisms of different second primary cancers require further investigation
Different FPCs may tend to develop different types of SPCs, and the mechanisms involved vary in different patterns. Further research is imperative to elucidate the etiology and mechanisms of different patterns.
Previous studies have analyzed the potential correlation between SPC occurrence and FPC, and explored the possible etiology and mechanisms of their patterns [5]. For instance, patients with first primary cancers occurring in four gynecological organs––namely the fallopian tube, uterus, vulva, vagina, and thyroid are more prone to develop second primary breast cancer and colorectal cancer. This phenomenon may be attributed to genetic susceptibility, the effect of hormone and radiation therapy after the FPC. Additionally, individuals with primary bladder or primary penile cancers have an increased likelihood of developing second primary prostate or lung cancer, which could be linked to their N-acetyltransferase genotype and smoking history. Furthermore, a high frequency of second primary bladder cancer is observed in patients with first primary ureteral cancer. This may be related to the clonal origin of upper urinary tract urothelial carcinoma, intraluminal seeding, and the treatment of ureteral cancer.
Additionally, a comprehensive monograph has analyzed the risk of subsequent cancers based on the primary site of cancer, providing insights into the potential etiology and risk factors involved [35].
Our study augments the existing knowledge on the etiology and mechanisms of various multiple primary cancers: ① During the subsequent development of second primary epithelial cancer in patients with head and neck cancer, elevated IGF-1 levels may contribute significantly. ② In the pattern of concurrent thyroid cancer and breast cancer, high levels of estrogen or overexpression of estrogen receptors (ER) may serve as contributing factors. ③ The development of second primary non-Hodgkin lymphoma (NHL) involves immunosuppression caused by the FPC. ④ In the pattern where breast cancer and lung cancer can mutually occur as each other's second primary cancer, EGFR mutations play a significant role.
Preventing SSB accumulation-induced senescence SPCs after FPC radiotherapy
SSB accumulation from off-target doses during FPC radiotherapy induces senescence and subsequent carcinogenesis, presenting a possible SPC mechanism and prevention opportunity. First, supplementing NAD + dietary precursors and modulating NAD + biosynthesis could prevent therapy edge senescence [206, 207], as PARylation uses NAD + to repair SSBs and maintain DNA integrity [87, 208]. Second, senolytic drugs to clear senescent cells at radiotherapy end may prevent senescence-induced SPCs [209,210,211].
Further elucidating IGF-1's SPC role and predicting SPCs via IGF-1 levels
While studies show IGF-1 promotes proliferation, inhibits apoptosis, and stimulates angiogenesis in carcinogenesis [135,136,137,138,139,140,141], some highlight uncertainty around IGF-1's SPC role [133]. More observational research is needed to clarify IGF-1's contributions to specific SPCs and involved IGF-1R downstream pathways. Though IGF-1's SPC mechanisms remain unclear, elevated levels associate with SPC risk. Additional studies could thus assess serum IGF-1's SPC risk predictive utility, with clinical monitoring and early detection implications.
Investigating common risk factors in SPC development
High estrogen levels play carcinogenic roles in thyroid and breast cancers [152,153,154, 156, 157, 212, 213], presenting a potential common risk factor for both cancers. Although some studies speculate shared hormone receptors and pathways may enable thyroid-breast cancer co-occurrence [167], no evidence shows the first primary cancer alters estrogen to cause the second primary in the other organ. Similarly, while EGFR overexpression associates with breast and lung cancers [185], no evidence demonstrates directional carcinogenesis between lung and breast primaries via EGFR. Elucidating whether common risk factors play exact roles in cancer concurrences requires further SPC mechanism studies.
Intratumoral microbes in SPC
Cancer intratumoral microbiota may originate from mucosa, neighboring tissues, circulation, and the gut [214]. Intratumoral microbes promote carcinogenesis through DNA damage, mutation, oncogenic pathway activation, and tumor microenvironment influence [214]. Beyond its role in cancer development, microbiota also regulate therapeutic efficacy, providing potential microbiota modulation-based strategies [214]. However, the microbiota-SPC relationship remains unclear. Questions include: do SPC microbes originate similarly? Do microbes also drive SPCs through these mechanisms? Can SPC microbes be specifically regulated without disturbing other body microbiota in application to the treatment of SPC? Intratumoral microbes are highly heterogeneous in different tumor types and specific microbial metabolic pathways correlate with specific tumor types [215], so what role might Intratumoral microbial diversity play in the development of different SPCs?
Single-cell multi-omics for SPC mechanisms
Single-cell multi-omics analyses of genomes, transcriptomes, epigenomes and proteomes overcome batch limitations and enable detailed study of cancer cell/molecular biology. Single-cell cancer studies have resolved heterogeneity, microenvironment interactions, and evolutionary details [216], suggesting single-cell multi-omics applications in SPC. However, SPC mechanism data based on single-cell multi-omics is lacking; more single-cell omics SPC projects are needed. Key questions include: Do certain SPC mutations or pathway activations differ from corresponding primary cancers at the molecular level, identifiable via single-cell multi-omics? If so, personalized SPC treatments may be possible.
Metabolomics for SPC mechanisms and screening
A non-targeted metabolomics study identified higher 5-MTA, phenylacetylglutamine, and valylglycine levels in second versus first primary lung cancers [217]. Though associated with various primary cancer risks [218,219,220,221], links to SPC mechanisms need elucidation. However, metabolomics shows promise for SPC mechanism exploration and screening. Key questions: (1) Can metabolomics determine causal metabolite-SPC links? (2) The metabolic transformation observed in different tumor processes is heterogeneous [222], so whether the expression patterns of metabolites are different in different SPC? Can we predict SPC by monitoring the levels of certain metabolites in the blood? Do different SPCs show heterogeneous metabolic patterns enabling screening via blood metabolites? (3) Can blocking SPC-related metabolites in FPC patients prevent SPCs? (4) Are protective metabolite changes known? (5) Do immune cell metabolic aberrations also drive SPCs? (6) Do certain metabolites confer SPC treatment resistance and poorer outcomes?
Conclusion
Second primary cancers (SPCs) pose a serious global public health threat that endangers first primary cancer (FPC) survivors. While researchers have intensively studied SPC mechanisms in recent years, regarding exact causes many questions remain. This review synthesizes factors potentially influencing SPC development, including genomic changes, stromal cell phenotypic/metabolic changes, immunosuppression, hormonal changes, aberrant gene methylation, EGFR signaling, and circulating DNA. These factors may individually or jointly affect SPCs. However, their specific roles across various SPC types and potential interactions warrant further research. We also explored emerging fields like intratumoral microbiology, single-cell multi-omics, and metabolomics as promising approaches for elucidating SPC mechanisms. These cutting-edge technologies and approaches have the potential to provide deeper, more comprehensive SPC mechanism insights, thereby informing preventive and therapeutic advancements (Fig. 4).
Mechanisms of SPC occurrence and future research directions. SPC mechanisms include genomic changes caused by treatment for FPC, cancer-associated fibroblast (CAF) induced by chemotherapy, first primary cancer (FPC)-induced immunosuppression, abnormal gene methylation, signaling pathway activation, and FPC-released cell-free DNA
Overall, SPC research still faces many challenges. Nevertheless,continued scientific and technological advancements, coupled with rigorous investigation, are expected to deepen our understanding of SPCs, potentially enabling more effective prevention strategies and treatment. Future research should aim to further elucidate specific factor roles in SPC development and their complex interrelationships. Simultaneously, more sensitive and specific methods for early SPC detection and innovative prevention strategies must be developed and validated. Through persistent and collaborative efforts, we can unravel the complex mechanisms underlying SPCs and translate these findings into life-saving outcomes for FPC survivors threatened by these devastating secondary malignancies.
Availability of data and materials
Data sharing not applicable – no new data generated, or the article describes entirely theoretical research.
Abbreviations
- SPCs:
-
Second primary cancers
- FPC:
-
First primary cancer
- SSBs:
-
Single-strand breaks
- PTV:
-
Planning target volume
- PARylation:
-
Poly(ADP-ribosyl)ation
- DSBs:
-
Double-strand breaks
- PSNE:
-
Post-senescence neoplastic emergence
- EMT:
-
Epithelial-mesenchymal transition
- hTERT:
-
Human telomerase reverse transcriptase
- B/F/B:
-
Breakage-fusion-bridge
- SNPs:
-
Single nucleotide polymorphisms
- HNSCC:
-
Head and neck squamous cell carcinoma
- CAF:
-
Cancer-associated fibroblast
- IGF-1:
-
Insulin-like growth factor 1
- VEGF:
-
Vascular endothelial growth factor
- GH:
-
Growth hormone
- ER:
-
Estrogen receptor
- MAPK:
-
Mitogen-activated protein kinase
- NHL:
-
Non-Hodgkin lymphoma
- EBV:
-
Epstein-Barr virus
- uPA:
-
Urokinase-type plasminogen activator
- ECM:
-
Extracellular matrix
- SPC:
-
Second primary cancer
- LVL:
-
Lugol-voiding lesion
- ESCC:
-
Esophageal squamous cell carcinoma
- ALDH2:
-
Aldehyde dehydrogenase 2
- TIMP3:
-
Tissue inhibitor of metalloproteinase 3
- MMP:
-
Matrix metalloproteinase
- cf-DNA:
-
Cell-free DNA
References
Vogt A, Schmid S, Heinimann K, Frick H, Herrmann C, Cerny T, et al. Multiple primary tumours: challenges and approaches, a review. ESMO Open. 2017;2(2): e000172.
Okeke F, Nriagu VC, Nwaneki CM, Magacha HM, Omenuko NJ, Anazor S. Factors that determine multiple primary cancers in the adult population in the United States. Cureus. 2023;15(9): e44993.
Murphy CC, Gerber DE, Pruitt SL. Prevalence of prior cancer among persons newly diagnosed with cancer: an initial report from the surveillance, epidemiology, and end results program. JAMA Oncol. 2018;4(6):832–6.
Qiao B, Hsieh MC, Wu XC, Kruchko C, Sineshaw H, Wu M, et al. Multiple primary cancers in the United States. J Registry Manag. 2020;47(2):60–6.
Wang X, Zeng M, Ju X, Lin A, Zhou C, Shen J, et al. Correlation between second and first primary cancer: systematic review and meta-analysis of 9 million cancer patients. Bri J Surgery. 2024;111(1):377.
Chao C, Bhatia S, Xu L, Cannavale KL, Wong FL, Huang PYS, et al. Incidence, risk factors, and mortality associated with second malignant neoplasms among survivors of adolescent and young adult cancer. JAMA Netw Open. 2019;2(6): e195536.
Macq G, Silversmit G, Verdoodt F, Van Eycken L. The epidemiology of multiple primary cancers in Belgium (2004–2017): Incidence, proportion, risk, stage and impact on relative survival estimates. BMC Cancer. 2023;23(1):349.
Curtis RE, Freedman DM, Ron E, Ries LAG, Hacker DG, Edwards BK, Tucker MA, Fraumeni JF Jr. New Malignancies Among Cancer Survivors SEER Cancer Registries, 1973–2000.
Sung H, Hyun N, Leach CR, Yabroff KR, Jemal A. Association of first primary cancer with risk of subsequent primary cancer among survivors of adult-onset cancers in the United States. JAMA. 2020;324(24):2521–35.
Oeffinger KC, Baxi SS, Novetsky Friedman D, Moskowitz CS. Solid tumor second primary neoplasms: who is at risk, what can we do? Semin Oncol. 2013;40(6):676–89.
Zhang S, Xiao X, Yi Y, Wang X, Zhu L, Shen Y, et al. Tumor initiation and early tumorigenesis: molecular mechanisms and interventional targets. Signal Transduct Target Ther. 2024;9(1):149.
Seeler JS, Dejean A. SUMO and the robustness of cancer. Nat Rev Cancer. 2017;17(3):184–97.
Bugter JM, Fenderico N, Maurice MM. Mutations and mechanisms of WNT pathway tumour suppressors in cancer. Nat Rev Cancer. 2021;21(1):5–21.
Dawson MA, Kouzarides T. Cancer epigenetics: from mechanism to therapy. Cell. 2012;150(1):12–27.
Parsons MJ, Tammela T, Dow LE. WNT as a driver and dependency in cancer. Cancer Discov. 2021;11(10):2413–29.
Martínez-Reyes I, Chandel NS. Cancer metabolism: looking forward. Nat Rev Cancer. 2021;21(10):669–80.
Wang T, Gao Y. Metabolic insights into tumor pathogenesis: unveiling pan-cancer metabolism and the potential of untargeted metabolomics. MedComm Future Med. 2023;2(3): e59.
Guo J, Zhao Z, Shang Z, Tang Z, Zhu H, Zhang K. Nanodrugs with intrinsic radioprotective exertion: Turning the double-edged sword into a single-edged knife. Exploration. 2023;3(2):20220119.
Najafi M, Salajegheh A, Rezaeyan A. Bystander effect and second primary cancers following radiotherapy: what are its significances? J Med Phys. 2017;42(1):55–6.
Najafi M, Fardid R, Hadadi G, Fardid M. The mechanisms of radiation-induced bystander effect. J Biomed Phys Eng. 2014;4(4):163–72.
Zhang M, Zou Q. Determination of two potential genotoxic impurities in crisaborole APIby UPLC-MS/MS. CPA. 2023;19(6):511–20.
Liu SZ, Jin SZ, Liu XD. Radiation-induced bystander effect in immune response. Biomed Environ Sci. 2004;17(1):40–6.
Zamarron BF, Chen W. Dual roles of immune cells and their factors in cancer development and progression. Int J Biol Sci. 2011;7(5):651–8.
Lakshmi Narendra B, Eshvendar Reddy K, Shantikumar S, Ramakrishna S. Immune system: a double-edged sword in cancer. Inflamm Res. 2013;62(9):823–34.
Ding Y, Wang Y, Hu Q. Recent advances in overcoming barriers to cell-based delivery systems for cancer immunotherapy. Exploration. 2022;2(3):20210106.
Peng S, Lin A, Jiang A, Zhang C, Zhang J, Cheng Q, et al. CTLs heterogeneity and plasticity: implications for cancer immunotherapy. Mol Cancer. 2024;23(1):58.
Li Y, Zhang C, Jiang A, Lin A, Liu Z, Cheng X, et al. Potential anti-tumor effects of regulatory T cells in the tumor microenvironment: a review. J Transl Med. 2024;22(1):293.
Gao Z, Bai Y, Lin A, Jiang A, Zhou C, Cheng Q, et al. Gamma delta T-cell-based immune checkpoint therapy: attractive candidate for antitumor treatment. Mol Cancer. 2023;22(1):31.
Travis LB, Rabkin CS, Brown LM, Allan JM, Alter BP, Ambrosone CB, et al. Cancer survivorship–genetic susceptibility and second primary cancers: research strategies and recommendations. J Natl Cancer Inst. 2006;98(1):15–25.
Birch JM, Hartley AL, Tricker KJ, Prosser J, Condie A, Kelsey AM, et al. Prevalence and diversity of constitutional mutations in the p53 gene among 21 Li-Fraumeni families. Cancer Res. 1994;54(5):1298–304.
Gonzalez KD, Noltner KA, Buzin CH, Gu D, Wen-Fong CY, Nguyen VQ, et al. Beyond Li Fraumeni Syndrome: clinical characteristics of families with p53 germline mutations. J Clin Oncol. 2009;27(8):1250–6.
Olivier M, Goldgar DE, Sodha N, Ohgaki H, Kleihues P, Hainaut P, et al. Li-Fraumeni and related syndromes: correlation between tumor type, family structure, and TP53 genotype. Cancer Res. 2003;63(20):6643–50.
Liu Y, Wang W, Zhang D, Sun Y, Li F, Zheng M, et al. Brain co-delivery of first-line chemotherapy drug and epigenetic bromodomain inhibitor for multidimensional enhanced synergistic glioblastoma therapy. Exploration. 2022;2(4):20210274.
Zhang Z, Huang J, Xu Y, Luo H. Chemoimmunotherapy for esophageal squamous cell carcinoma—Summary and discussion of recent clinical trials. MedComm Future Med. 2023;2(3): e56.
Supramaniam R. New malignancies among cancer survivors: SEER cancer registries, 1973–2000. J Epidemiol Community Health. 2008;62(4):375–6.
Du B, Leung H, Khan KMF, Miller CG, Subbaramaiah K, Falcone DJ, et al. Tobacco smoke induces urokinase-type plasminogen activator and cell invasiveness: evidence for an epidermal growth factor receptor dependent mechanism. Cancer Res. 2007;67(18):8966–72.
Khuri FR, Kim ES, Lee JJ, Winn RJ, Benner SE, Lippman SM, et al. The impact of smoking status, disease stage, and index tumor site on second primary tumor incidence and tumor recurrence in the head and neck retinoid chemoprevention trial. Cancer Epidemiol Biomarkers Prev. 2001;10(8):823–9.
Petersen LØ, Jensen JS, Jakobsen KK, Grønhøj C, Wessel I, von Buchwald C. Second primary cancer following primary oral squamous cell carcinoma: a population-based, retrospective study. Acta Oncol. 2022;61(8):916–21.
Do KA, Johnson MM, Doherty DA, Lee JJ, Wu XF, Dong Q, et al. Second primary tumors in patients with upper aerodigestive tract cancers: joint effects of smoking and alcohol (United States). Cancer Causes Control. 2003;14(2):131–8.
Du B, Altorki NK, Kopelovich L, Subbaramaiah K, Dannenberg AJ. Tobacco smoke stimulates the transcription of amphiregulin in human oral epithelial cells: evidence of a cyclic AMP-responsive element binding protein-dependent mechanism. Cancer Res. 2005;65(13):5982–8.
Alessi DR, Cuenda A, Cohen P, Dudley DT, Saltiel AR. PD 098059 is a specific inhibitor of the activation of mitogen-activated protein kinase kinase in vitro and in vivo. J Biol Chem. 1995;270(46):27489–94.
Mahabeleshwar GH, Kundu GC. Tyrosine kinase p56lck regulates cell motility and nuclear factor kappaB-mediated secretion of urokinase type plasminogen activator through tyrosine phosphorylation of IkappaBalpha following hypoxia/reoxygenation. J Biol Chem. 2003;278(52):52598–612.
Henkin J, Marcotte P, Yang HC. The plasminogen-plasmin system. Prog Cardiovasc Dis. 1991;34(2):135–64.
Moser TL, Enghild JJ, Pizzo SV, Stack MS. The extracellular matrix proteins laminin and fibronectin contain binding domains for human plasminogen and tissue plasminogen activator. J Biol Chem. 1993;268(25):18917–23.
Pavón MA, Arroyo-Solera I, Céspedes MV, Casanova I, León X, Mangues R. uPA/uPAR and SERPINE1 in head and neck cancer: role in tumor resistance, metastasis, prognosis and therapy. Oncotarget. 2016;7(35):57351–66.
Andreasen PA, Kjøller L, Christensen L, Duffy MJ. The urokinase-type plasminogen activator system in cancer metastasis: a review. Int J Cancer. 1997;72(1):1–22.
Mao L, Hong WK, Papadimitrakopoulou VA. Focus on head and neck cancer. Cancer Cell. 2004;5(4):311–6.
Jang SJ, Chiba I, Hirai A, Hong WK, Mao L. Multiple oral squamous epithelial lesions: are they genetically related? Oncogene. 2001;20(18):2235–42.
Califano J, Leong PL, Koch WM, Eisenberger CF, Sidransky D, Westra WH. Second esophageal tumors in patients with head and neck squamous cell carcinoma: an assessment of clonal relationships. Clin Cancer Res. 1999;5(7):1862–7.
Jo M, Thomas KS, O’Donnell DM, Gonias SL. Epidermal growth factor receptor-dependent and -independent cell-signaling pathways originating from the urokinase receptor. J Biol Chem. 2003;278(3):1642–6.
Inoue H, Rey JF, Lightdale C. Lugol chromoendoscopy for esophageal squamous cell cancer. Endoscopy. 2001;33(1):75–9.
Katada C, Muto M, Tanabe S, Higuchi K, Sasaki T, Azuma M, et al. Factors associated with the presence of multiple Lugol-voiding lesions in patients with esophageal squamous-cell carcinoma. Dis Esophagus. 2014;27(5):457–62.
Muto M, Takahashi M, Ohtsu A, Ebihara S, Yoshida S, Esumi H. Risk of multiple squamous cell carcinomas both in the esophagus and the head and neck region. Carcinogenesis. 2005;26(5):1008–12.
Muto M, Nakane M, Hitomi Y, Yoshida S, Sasaki S, Ohtsu A, et al. Association between aldehyde dehydrogenase gene polymorphisms and the phenomenon of field cancerization in patients with head and neck cancer. Carcinogenesis. 2002;23(10):1759–65.
Priante AVM, Castilho EC, Kowalski LP. Second primary tumors in patients with head and neck cancer. Curr Oncol Rep. 2011;13(2):132–7.
Licciardello JT, Spitz MR, Hong WK. Multiple primary cancer in patients with cancer of the head and neck: second cancer of the head and neck, esophagus, and lung. Int J Radiat Oncol Biol Phys. 1989;17(3):467–76.
Katada C, Yokoyama T, Yano T, Kaneko K, Oda I, Shimizu Y, et al. Alcohol consumption and multiple dysplastic lesions increase risk of squamous cell carcinoma in the esophagus, head, and neck. Gastroenterology. 2016;151(5):860-869.e7.
Hori K, Okada H, Kawahara Y, Takenaka R, Shimizu S, Ohno Y, et al. Lugol-voiding lesions are an important risk factor for a second primary squamous cell carcinoma in patients with esosphageal cancer or head and neck cancer. Am J Gastroenterol. 2011;106(5):858–66.
Yokoyama A, Hirota T, Omori T, Yokoyama T, Kawakubo H, Matsui T, et al. Development of squamous neoplasia in esophageal iodine-unstained lesions and the alcohol and aldehyde dehydrogenase genotypes of Japanese alcoholic men. Int J Cancer. 2012;130(12):2949–60.
Muto M, Hironaka S, Nakane M, Boku N, Ohtsu A, Yoshida S. Association of multiple Lugol-voiding lesions with synchronous and metachronous esophageal squamous cell carcinoma in patients with head and neck cancer. Gastrointest Endosc. 2002;56(4):517–21.
Urabe Y, Hiyama T, Tanaka S, Oka S, Yoshihara M, Arihiro K, et al. Metachronous multiple esophageal squamous cell carcinomas and Lugol-voiding lesions after endoscopic mucosal resection. Endoscopy. 2009;41(4):304–9.
Bevers TB. Screening for Second Primary Cancers. In: Foxhall LE, Rodriguez MA, editors. Advances in Cancer Survivorship Management. Springer, New York: New York NY; 2015. p. 299–321.
Day GL, Shore RE, Blot WJ, McLaughlin JK, Austin DF, Greenberg RS, et al. Dietary factors and second primary cancers: a follow-up of oral and pharyngeal cancer patients. Nutr Cancer. 1994;21(3):223–32.
Nisha P, Srushti P, Bhavarth D, Kaif M, Palak P. Comprehensive review on analytical and bioanalytical methods for quantification of anti-angiogenic agents used in treatment of cervicalcancer. CPA. 2023;19(10):735–44.
Umar A, Boland CR, Terdiman JP, Syngal S, de la Chapelle A, Rüschoff J, et al. Revised Bethesda Guidelines for hereditary nonpolyposis colorectal cancer (Lynch syndrome) and microsatellite instability. J Natl Cancer Inst. 2004;96(4):261–8.
Cavazos TB, Kachuri L, Graff RE, Nierenberg JL, Thai KK, Alexeeff S, et al. Assessment of genetic susceptibility to multiple primary cancers through whole-exome sequencing in two large multi-ancestry studies. BMC Med. 2022;20(1):332.
Bakos B, Kiss A, Árvai K, Szili B, Deák-Kocsis B, Tobiás B, et al. Co-occurrence of thyroid and breast cancer is associated with an increased oncogenic SNP burden. BMC Cancer. 2021;21(1):706.
Griffith TS, Brunner T, Fletcher SM, Green DR, Ferguson TA. Fas ligand-induced apoptosis as a mechanism of immune privilege. Science. 1995;270(5239):1189–92.
Reichmann E. The biological role of the Fas/FasL system during tumor formation and progression. Semin Cancer Biol. 2002;12(4):309–15.
Zhang Z, Wang LE, Sturgis EM, El-Naggar AK, Hong WK, Amos CI, et al. Polymorphisms of FAS and FAS ligand genes involved in the death pathway and risk and progression of squamous cell carcinoma of the head and neck. Clin Cancer Res. 2006;12(18):5596–602.
Ueda M, Terai Y, Kanda K, Kanemura M, Takehara M, Yamaguchi H, et al. Fas gene promoter -670 polymorphism in gynecological cancer. Int J Gynecol Cancer. 2006;16(Suppl 1):179–82.
Sun T, Zhou Y, Li H, Han X, Shi Y, Wang L, et al. FASL -844C polymorphism is associated with increased activation-induced T cell death and risk of cervical cancer. J Exp Med. 2005;202(7):967–74.
Cao Y, Miao XP, Huang MY, Deng L, Lin DX, Zeng YX, et al. Polymorphisms of death pathway genes FAS and FASL and risk of nasopharyngeal carcinoma. Mol Carcinog. 2010;49(11):944–50.
Lei D, Sturgis EM, Wang LE, Liu Z, Zafereo ME, Wei Q, et al. FAS and FASLG genetic variants and risk for second primary malignancy in patients with squamous cell carcinoma of the head and neck. Cancer Epidemiol Biomark Prev. 2010;19(6):1484–91.
Wang Z, Sturgis EM, Zhang F, Lei D, Liu Z, Xu L, et al. Genetic variants of p27 and p21 as predictors for risk of second primary malignancy in patients with index squamous cell carcinoma of head and neck. Mol Cancer. 2012;26(11):17.
Lei D, Sturgis EM, Liu Z, Zafereo ME, Wei Q, Li G. Genetic polymorphisms of p21 and risk of second primary malignancy in patients with index squamous cell carcinoma of the head and neck. Carcinogenesis. 2010;31(2):222–7.
Hoxhaj I, Vukovic V, Boccia S, Pastorino R. Single nucleotide polymorphisms and the risk of developing a second primary cancer among head and neck cancer patients: a systematic literature review and meta-analysis. BMC Cancer. 2021;21(1):660.
Hubert JN, Suybeng V, Vallée M, Delhomme TM, Maubec E, Boland A, et al. The PI3K/mTOR pathway is targeted by rare germline variants in patients with both melanoma and renal cell carcinoma. Cancers (Basel). 2021;13(9):2243.
Jiang A, Xu Z, Fang X, Gu D, Dong K, Wu Z, et al. RNA modification pattern-based subtypes reveal heterogenous clinical outcomes and tumor immunity of clear cell renal cell carcinoma. MedComm Future Med. 2023;2(1): e30.
Lin A, Gu T, Hu X, Zhang J, Luo P. Comprehensive analysis identifies PI3K/Akt pathway alternations as an immune-related prognostic biomarker in colon adenocarcinoma patients receiving immune checkpoint inhibitor treatment. J Immunol Res. 2022;2022:8179799.
Koubková L, Hrstka R, Dobeš P, Vojtěšek B, Vyzula R. Second primary cancers—causes. Incidence and the Future Klin Onkol. 2014;27(1):11–7.
Travis LB, Hill DA, Dores GM, Gospodarowicz M, Van Leeuwen FE, Holowaty E, et al. Breast cancer following radiotherapy and chemotherapy among young women with hodgkin disease. JAMA. 2003;290(4):465.
Goy E, Tomezak M, Facchin C, Martin N, Bouchaert E, Benoit J, et al. The out-of-field dose in radiation therapy induces delayed tumorigenesis by senescence evasion. Elife. 2022;18(11): e67190.
Chofor N, Harder D, Willborn KC, Poppe B. Internal scatter, the unavoidable major component of the peripheral dose in photon-beam radiotherapy. Phys Med Biol. 2012;57(6):1733–43.
Reisz JA, Bansal N, Qian J, Zhao W, Furdui CM. Effects of ionizing radiation on biological molecules–mechanisms of damage and emerging methods of detection. Antioxid Redox Signal. 2014;21(2):260–92.
D’Amours D, Desnoyers S, D’Silva I, Poirier GG. Poly(ADP-ribosyl)ation reactions in the regulation of nuclear functions. Biochem J. 1999;342:249–68.
Bürkle A, Virág L. Poly(ADP-ribose): PARadigms and PARadoxes. Mol Aspects Med. 2013;34(6):1046–65.
Fang EF, Scheibye-Knudsen M, Brace LE, Kassahun H, SenGupta T, Nilsen H, et al. Defective mitophagy in XPA via PARP-1 hyperactivation and NAD(+)/SIRT1 reduction. Cell. 2014;157(4):882–96.
Cmielová J, Havelek R, Jiroutová A, Kohlerová R, Seifrtová M, Muthná D, et al. DNA damage caused by ionizing radiation in embryonic diploid fibroblasts WI-38 induces both apoptosis and senescence. Physiol Res. 2011;60(4):667–77.
Nassour J, Martien S, Martin N, Deruy E, Tomellini E, Malaquin N, et al. Defective DNA single-strand break repair is responsible for senescence and neoplastic escape of epithelial cells. Nat Commun. 2016;7(1):10399.
Rayess H, Wang MB, Srivatsan ES. Cellular senescence and tumor suppressor gene p16. Int J Cancer. 2012;130(8):1715–25.
Azzam EI, Jay-Gerin JP, Pain D. Ionizing radiation-induced metabolic oxidative stress and prolonged cell injury. Cancer Lett. 2012;327(1–2):48–60.
Jonathan EC, Bernhard EJ, McKenna WG. How does radiation kill cells? Curr Opin Chem Biol. 1999;3(1):77–83.
Panganiban RAM, Snow AL, Day RM. Mechanisms of radiation toxicity in transformed and non-transformed cells. Int J Mol Sci. 2013;14(8):15931–58.
Shi C, Qin K, Lin A, Jiang A, Cheng Q, Liu Z, et al. The role of DNA damage repair (DDR) system in response to immune checkpoint inhibitor (ICI) therapy. J Exp Clin Cancer Res. 2022;41(1):268.
Zhou C, Lin A, Cao M, Ding W, Mou W, Guo N, et al. Activation of the DDR Pathway leads to the down-regulation of the TGFβ Pathway and a better response to icis in patients with metastatic urothelial carcinoma. Front Immunol. 2021;12: 634741.
Luo P, Lin A, Li K, Wei T, Zhang J. DDR pathway alteration, tumor mutation burden, and cisplatin sensitivity in small cell lung cancer: difference detected by whole exome and targeted gene sequencing. J Thorac Oncol. 2019;14(12):e276–9.
Caldecott KW. Single-strand break repair and genetic disease. Nat Rev Genet. 2008;9(8):619–31.
Gosselin K, Martien S, Pourtier A, Vercamer C, Ostoich P, Morat L, et al. Senescence-associated oxidative DNA damage promotes the generation of neoplastic cells. Can Res. 2009;69(20):7917–25.
Krtolica A, Parrinello S, Lockett S, Desprez PY, Campisi J. Senescent fibroblasts promote epithelial cell growth and tumorigenesis: a link between cancer and aging. Proc Natl Acad Sci U S A. 2001;98(21):12072–7.
Martin N, Salazar-Cardozo C, Vercamer C, Ott L, Marot G, Slijepcevic P, et al. Identification of a gene signature of a pre-transformation process by senescence evasion in normal human epidermal keratinocytes. Mol Cancer. 2014;14(13):151.
Malaquin N, Vercamer C, Bouali F, Martien S, Deruy E, Wernert N, et al. Senescent fibroblasts enhance early skin carcinogenic events via a paracrine MMP-PAR-1 axis. PLoS ONE. 2013;8(5): e63607.
Su S, Lin A, Luo P, Zou J, Huang Z, Wang X, et al. Effect of mesenchymal-epithelial transition amplification on immune microenvironment and efficacy of immune checkpoint inhibitors in patients with non-small cell lung cancer. Ann Transl Med. 2021;9(18):1475.
Gramatges MM, Liu Q, Yasui Y, Okcu MF, Neglia JP, Strong LC, et al. Telomere content and risk of second malignant neoplasm in survivors of childhood cancer: a report from the Childhood Cancer Survivor Study. Clin Cancer Res. 2014;20(4):904–11.
Li P, Hou M, Lou F, Björkholm M, Xu D. Telomere dysfunction induced by chemotherapeutic agents and radiation in normal human cells. Int J Biochem Cell Biol. 2012;44(9):1531–40.
Schröder CP, Wisman GB, de Jong S, van der Graaf WT, Ruiters MH, Mulder NH, et al. Telomere length in breast cancer patients before and after chemotherapy with or without stem cell transplantation. Br J Cancer. 2001;84(10):1348–53.
Beeharry N, Broccoli D. Telomere dynamics in response to chemotherapy. Curr Mol Med. 2005;5(2):187–96.
Guidetti A, Carlo-Stella C, Ruella M, Miceli R, Devizzi L, Locatelli SL, et al. Myeloablative doses of yttrium-90-ibritumomab tiuxetan and the risk of secondary myelodysplasia/acute myelogenous leukemia. Cancer. 2011;117(22):5074–84.
Ilyenko I, Lyaskivska O, Bazyka D. Analysis of relative telomere length and apoptosis in humans exposed to ionising radiation. Exp Oncol. 2011;33(4):235–8.
Murnane JP. Telomeres and chromosome instability. DNA Repair. 2006;5(9–10):1082–92.
Montgomery E, Wilentz RE, Argani P, Fisher C, Hruban RH, Kern SE, et al. Analysis of anaphase figures in routine histologic sections distinguishes chromosomally unstable from chromosomally stable malignancies. Cancer Biol Ther. 2003;2(3):248–52.
Murnane JP. Telomere dysfunction and chromosome instability. Mutation Res/Fund Mol Mechan Mutagenesis. 2012;730(1–2):28–36.
Lengauer C, Kinzler KW, Vogelstein B. Genetic instabilities in human cancers. Nature. 1998;396(6712):643–9.
Watkins TBK, Lim EL, Petkovic M, Elizalde S, Birkbak NJ, Wilson GA, et al. Pervasive chromosomal instability and karyotype order in tumour evolution. Nature. 2020;587(7832):126–32.
Ford M, Fried M. Large inverted duplications are associated with gene amplification. Cell. 1986;45(3):425–30.
Campbell PJ, Yachida S, Mudie LJ, Stephens PJ, Pleasance ED, Stebbings LA, et al. The patterns and dynamics of genomic instability in metastatic pancreatic cancer. Nature. 2010;467(7319):1109–13.
Tanaka H, Bergstrom DA, Yao MC, Tapscott SJ. Widespread and nonrandom distribution of DNA palindromes in cancer cells provides a structural platform for subsequent gene amplification. Nat Genet. 2005;37(3):320–7.
Svetec Miklenić M, Svetec IK. Palindromes in DNA-A Risk for Genome Stability and Implications in Cancer. Int J Mol Sci. 2021;22(6):2840.
Wu X, Amos CI, Zhu Y, Zhao H, Grossman BH, Shay JW, et al. Telomere dysfunction: a potential cancer predisposition factor. J Natl Cancer Inst. 2003;95(16):1211–8.
Cantara S, Pisu M, Frau DV, Caria P, Dettori T, Capezzone M, et al. Telomere abnormalities and chromosome fragility in patients affected by familial papillary thyroid cancer. J Clin Endocrinol Metab. 2012;97(7):E1327-1331.
Capezzone M, Cantara S, Marchisotta S, Filetti S, De Santi MM, Rossi B, et al. Short telomeres, telomerase reverse transcriptase gene amplification, and increased telomerase activity in the blood of familial papillary thyroid cancer patients. J Clin Endocrinol Metab. 2008;93(10):3950–7.
Peiris-Pagès M, Sotgia F, Lisanti MP. Chemotherapy induces the cancer-associated fibroblast phenotype, activating paracrine Hedgehog-GLI signalling in breast cancer cells. Oncotarget. 2015;6(13):10728–45.
Martinez-Outschoorn U, Sotgia F, Lisanti MP. Tumor microenvironment and metabolic synergy in breast cancers: critical importance of mitochondrial fuels and function. Semin Oncol. 2014;41(2):195–216.
Pavlides S, Tsirigos A, Migneco G, Whitaker-Menezes D, Chiavarina B, Flomenberg N, et al. The autophagic tumor stroma model of cancer: Role of oxidative stress and ketone production in fueling tumor cell metabolism. Cell Cycle. 2010;9(17):3485–505.
Toullec A, Gerald D, Despouy G, Bourachot B, Cardon M, Lefort S, et al. Oxidative stress promotes myofibroblast differentiation and tumour spreading. EMBO Mol Med. 2010;2(6):211–30.
Bonuccelli G, Tsirigos A, Whitaker-Menezes D, Pavlides S, Pestell RG, Chiavarina B, et al. Ketones and lactate ‘fuel’ tumor growth and metastasis: Evidence that epithelial cancer cells use oxidative mitochondrial metabolism. Cell Cycle. 2010;9(17):3506–14.
Pavlides S, Whitaker-Menezes D, Castello-Cros R, Flomenberg N, Witkiewicz AK, Frank PG, et al. The reverse Warburg effect: aerobic glycolysis in cancer associated fibroblasts and the tumor stroma. Cell Cycle. 2009;8(23):3984–4001.
Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008;454(7203):436–44.
Oshima H, Oshima M. The inflammatory network in the gastrointestinal tumor microenvironment: lessons from mouse models. J Gastroenterol. 2012;47(2):97–106.
Teglund S, Toftgård R. Hedgehog beyond medulloblastoma and basal cell carcinoma. Biochim Biophys Acta. 2010;1805(2):181–208.
Zhang Q, Raje V, Yakovlev VA, Yacoub A, Szczepanek K, Meier J, et al. Mitochondrial localized Stat3 promotes breast cancer growth via phosphorylation of serine 727. J Biol Chem. 2013;288(43):31280–8.
Sherwood V, Chaurasiya SK, Ekström EJ, Guilmain W, Liu Q, Koeck T, et al. WNT5A-mediated β-catenin-independent signalling is a novel regulator of cancer cell metabolism. Carcinogenesis. 2014;35(4):784–94.
Wu X, Zhao H, Do KA, Johnson MM, Dong Q, Hong WK, et al. Serum levels of insulin growth factor (IGF-I) and IGF-binding protein predict risk of second primary tumors in patients with head and neck cancer. Clin Cancer Res. 2004;10(12 Pt 1):3988–95.
Torrisi R, Decensi A, Formelli F, Camerini T, De Palo G. Chemoprevention of breast cancer with fenretinide. Drugs. 2001;61(7):909–18.
Kalfert D, Ludvikova M, Topolcan O, Celakovsky P, Kucera R, Windrichova J, et al. Serum Levels of IGF-1 and IGFBP-3 in Relation to Clinical and Pathobiological Aspects of Head and Neck Squamous Cell Carcinomas. Anticancer Res. 2017;37(6):3281–6.
Adams TE, Epa VC, Garrett TP, Ward CW. Structure and function of the type 1 insulin-like growth factor receptor. Cell Mol Life Sci. 2000;57(7):1050–93.
LeRoith D, Werner H, Beitner-Johnson D, Roberts CT. Molecular and cellular aspects of the insulin-like growth factor I receptor. Endocr Rev. 1995;16(2):143–63.
Bowers LW, Rossi EL, O’Flanagan CH, deGraffenried LA, Hursting SD. The role of the Insulin/IGF system in cancer: lessons learned from clinical trials and the energy balance-cancer link. Front Endocrinol (Lausanne). 2015;6:77.
Yamamura T, Otani H, Nakao Y, Hattori R, Osako M, Imamura H. IGF-I differentially regulates Bcl-xL and Bax and confers myocardial protection in the rat heart. Am J Physiol-Heart Circul Physiol. 2001;280(3):H1191–200.
Park SM, Lim MK, Jung KW, Shin SA, Yoo KY, Yun YH, et al. Prediagnosis smoking, obesity, insulin resistance, and second primary cancer risk in male cancer survivors: National Health Insurance Corporation Study. J Clin Oncol. 2007;25(30):4835–43.
Fukuda R, Hirota K, Fan F, Jung YD, Ellis LM, Semenza GL. Insulin-like growth factor 1 induces hypoxia-inducible factor 1-mediated vascular endothelial growth factor expression, which is dependent on MAP kinase and phosphatidylinositol 3-kinase signaling in colon cancer cells. J Biol Chem. 2002;277(41):38205–11.
Shanmugalingam T, Bosco C, Ridley AJ, Van Hemelrijck M. Is there a role for IGF-1 in the development of second primary cancers? Cancer Med. 2016;5(11):3353–67.
Hancock SL, Tucker MA, Hoppe RT. Breast cancer after treatment of Hodgkin’s disease. J Natl Cancer Inst. 1993;85(1):25–31.
Aisenberg AC, Finkelstein DM, Doppke KP, Koerner FC, Boivin JF, Willett CG. High risk of breast carcinoma after irradiation of young women with Hodgkin’s disease. Cancer. 1997;79(6):1203–10.
Kleinberg DL, Feldman M, Ruan W. IGF-I: an essential factor in terminal end bud formation and ductal morphogenesis. J Mammary Gland Biol Neoplasia. 2000;5(1):7–17.
Ruan W, Kleinberg DL. Insulin-like growth factor I is essential for terminal end bud formation and ductal morphogenesis during mammary development. Endocrinology. 1999;140(11):5075–81.
Kleinberg DL, Ruan W. IGF-I, GH, and sex steroid effects in normal mammary gland development. J Mammary Gland Biol Neoplasia. 2008;13(4):353–60.
Zhang D, Brodt P. Type 1 insulin-like growth factor regulates MT1-MMP synthesis and tumor invasion via PI 3-kinase/Akt signaling. Oncogene. 2003;22(7):974–82.
Frasca F, Pandini G, Sciacca L, Pezzino V, Squatrito S, Belfiore A, et al. The role of insulin receptors and IGF-I receptors in cancer and other diseases. Arch Physiol Biochem. 2008;114(1):23–37.
Smith J, Axelrod D, Singh B, Kleinberg D. Prevention of breast cancer: the case for studying inhibition of IGF-1 actions. Ann Oncol. 2011;22(Suppl 1):i50-52.
Vigneri PG, Tirrò E, Pennisi MS, Massimino M, Stella S, Romano C, et al. The Insulin/IGF system in colorectal cancer development and resistance to therapy. Front Oncol. 2015;5:230.
Colditz GA. Relationship between estrogen levels, use of hormone replacement therapy, and breast cancer. J Natl Cancer Inst. 1998;90(11):814–23.
Colditz GA, Hankinson SE, Hunter DJ, Willett WC, Manson JE, Stampfer MJ, et al. The use of estrogens and progestins and the risk of breast cancer in postmenopausal women. N Engl J Med. 1995;332(24):1589–93.
Collaborative Group on Hormonal Factors in Breast Cancer. Type and timing of menopausal hormone therapy and breast cancer risk: individual participant meta-analysis of the worldwide epidemiological evidence. Lancet. 2019;394(10204):1159–68.
Clusan L, Ferrière F, Flouriot G, Pakdel F. A basic review on estrogen receptor signaling pathways in breast cancer. Int J Mol Sci. 2023;24(7):6834.
Yane K, Kitahori Y, Konishi N, Okaichi K, Ohnishi T, Miyahara H, et al. Expression of the estrogen receptor in human thyroid neoplasms. Cancer Lett. 1994;84(1):59–66.
Kawabata W, Suzuki T, Moriya T, Fujimori K, Naganuma H, Inoue S, et al. Estrogen receptors (alpha and beta) and 17beta-hydroxysteroid dehydrogenase type 1 and 2 in thyroid disorders: possible in situ estrogen synthesis and actions. Mod Pathol. 2003;16(5):437–44.
Božović A, Mandušić V, Todorović L, Krajnović M. Estrogen receptor beta: the promising biomarker and potential target in metastases. Int J Mol Sci. 2021;22(4):1656.
Zeng Q, Chen GG, Vlantis AC, van Hasselt CA. Oestrogen mediates the growth of human thyroid carcinoma cells via an oestrogen receptor-ERK pathway. Cell Prolif. 2007;40(6):921–35.
Kumar A, Klinge CM, Goldstein RE. Estradiol-induced proliferation of papillary and follicular thyroid cancer cells is mediated by estrogen receptors alpha and beta. Int J Oncol. 2010;36(5):1067–80.
Manole D, Schildknecht B, Gosnell B, Adams E, Derwahl M. Estrogen promotes growth of human thyroid tumor cells by different molecular mechanisms. J Clin Endocrinol Metab. 2001;86(3):1072–7.
Liu J, Xu T, Ma L, Chang W. Signal pathway of Estrogen and Estrogen receptor in the development of thyroid cancer. Front Oncol. 2021;11: 593479.
Hima S, Sreeja S. Modulatory role of 17β-estradiol in the tumor microenvironment of thyroid cancer. IUBMB Life. 2016;68(2):85–96.
Kuo JH, Chabot JA, Lee JA. Breast cancer in thyroid cancer survivors: an analysis of the Surveillance, Epidemiology, and End Results-9 database. Surgery. 2016;159(1):23–9.
An JH, Hwangbo Y, Ahn HY, Keam B, Lee KE, Han W, et al. A possible association between thyroid cancer and breast cancer. Thyroid. 2015;25(12):1330–8.
Zhang L, Wu Y, Liu F, Fu L, Tong Z. Characteristics and survival of patients with metachronous or synchronous double primary malignancies: breast and thyroid cancer. Oncotarget. 2016;7(32):52450–9.
Kim YA, Kim YA, Cho SW, Song YS, Min HS, Park IA, et al. Increased expression of thyroid hormone receptor alpha and estrogen receptor alpha in breast cancer associated with thyroid cancer. Eur J Surg Oncol. 2021;47(6):1316–23.
Flamini MI, Uzair ID, Pennacchio GE, Neira FJ, Mondaca JM, Cuello-Carrión FD, et al. Thyroid hormone controls breast cancer cell movement via integrin αV/β3/SRC/FAK/PI3-Kinases. HORM CANC. 2017;8(1):16–27.
Wang D, Hu L, Zhang G, Zhang L, Chen C. G protein-coupled receptor 30 in tumor development. Endocrine. 2010;38(1):29–37.
Hernández-Silva CD, Villegas-Pineda JC, Pereira-Suárez AL. Expression and role of the G protein-coupled estrogen receptor (GPR30/GPER) in the development and immune response in female reproductive cancers. Front Endocrinol (Lausanne). 2020;11:544.
Fu J, He M, Wu Q, Zhang X, Qi X, Shen K, et al. The clinical and genetic features in patients coexisting primary breast and thyroid cancers. Front Endocrinol (Lausanne). 2023;14:1136120.
Shankland KR, Armitage JO, Hancock BW. Non-Hodgkin lymphoma. Lancet. 2012;380(9844):848–57.
Armitage JO, Gascoyne RD, Lunning MA, Cavalli F. Non-Hodgkin lymphoma. Lancet. 2017;390(10091):298–310.
Chattopadhyay S, Sud A, Zheng G, Yu H, Sundquist K, Sundquist J, et al. Second primary cancers in non-Hodgkin lymphoma: Bidirectional analyses suggesting role for immune dysfunction. Int J Cancer. 2018;143(10):2449–57.
Chattopadhyay S, Hemminki A, Försti A, Sundquist K, Sundquist J, Hemminki K. Second primary cancers in patients with invasive and in situ squamous cell skin carcinoma, kaposi sarcoma, and merkel cell carcinoma: role for immune mechanisms? J Invest Dermatol. 2020;140(1):48-55.e1.
Friman V, Winqvist O, Blimark C, Langerbeins P, Chapel H, Dhalla F. Secondary immunodeficiency in lymphoproliferative malignancies. Hematol Oncol. 2016;34(3):121–32.
Munn DH, Bronte V. Immune suppressive mechanisms in the tumor microenvironment. Curr Opin Immunol. 2016;39:1–6.
Hemminki K, Jiang Y, Steineck G. Skin cancer and non-Hodgkin’s lymphoma as second malignancies markers of impaired immune function? Eur J Cancer. 2003;39(2):223–9.
Rama I, Grinyó JM. Malignancy after renal transplantation: the role of immunosuppression. Nat Rev Nephrol. 2010;6(9):511–9.
Stetler-Stevenson WG, Aznavoorian S, Liotta LA. Tumor cell interactions with the extracellular matrix during invasion and metastasis. Annu Rev Cell Biol. 1993;9(1):541–73.
Gomez DE, Alonso DF, Yoshiji H, Thorgeirsson UP. Tissue inhibitors of metalloproteinases: structure, regulation and biological functions. Eur J Cell Biol. 1997;74(2):111–22.
Rettori MM, de Carvalho AC, Longo ALB, de Oliveira CZ, Kowalski LP, Carvalho AL, et al. TIMP3 and CCNA1 hypermethylation in HNSCC is associated with an increased incidence of second primary tumors. J Transl Med. 2013;20(11):316.
Sun W, Zaboli D, Wang H, Liu Y, Arnaoutakis D, Khan T, et al. Detection of TIMP3 promoter hypermethylation in salivary rinse as an independent predictor of local recurrence-free survival in head and neck cancer. Clin Cancer Res. 2012;18(4):1082–91.
de Carvalho AC, Kowalski LP, Campos AHJFM, Soares FA, Carvalho AL, Vettore AL. Clinical significance of molecular alterations in histologically negative surgical margins of head and neck cancer patients. Oral Oncol. 2012;48(3):240–8.
Zeng T, Xu H, Liu Y, Sun C, Yang F, Liang Y, et al. High rate of epidermal growth factor receptor-mutated primary lung cancer in patients with primary breast cancer. Front Oncol. 2022;12: 985734.
Wang R, Yin Z, Liu L, Gao W, Li W, Shu Y, et al. Second primary lung cancer after breast cancer: a population-based study of 6,269 women. Front Oncol. 2018;8:427.
Alanazi IO, Khan Z. Understanding EGFR signaling in breast cancer and breast cancer stem cells: overexpression and therapeutic implications. Asian Pac J Cancer Prev. 2016;17(2):445–53.
Levva S, Kotoula V, Kostopoulos I, Manousou K, Papadimitriou C, Papadopoulou K, et al. Prognostic evaluation of epidermal growth factor receptor (EGFR) genotype and phenotype parameters in triple-negative breast cancers. Cancer Genom Proteom. 2017;14(3):181–95.
Kondapaka SB, Fridman R, Reddy KB. Epidermal growth factor and amphiregulin up-regulate matrix metalloproteinase-9 (MMP-9) in human breast cancer cells. Int J Cancer. 1997;70(6):722–6.
Siegfried JM, Stabile LP. Estrongenic steroid hormones in lung cancer. Semin Oncol. 2014;41(1):5–16.
Stabile LP, Lyker JS, Gubish CT, Zhang W, Grandis JR, Siegfried JM. Combined targeting of the estrogen receptor and the epidermal growth factor receptor in non-small cell lung cancer shows enhanced antiproliferative effects. Cancer Res. 2005;65(4):1459–70.
Yarden Y, Sliwkowski MX. Untangling the ErbB signalling network. Nat Rev Mol Cell Biol. 2001;2(2):127–37.
De Luca A, Carotenuto A, Rachiglio A, Gallo M, Maiello MR, Aldinucci D, et al. The role of the EGFR signaling in tumor microenvironment. J Cell Physiol. 2008;214(3):559–67.
Larsen AK, Ouaret D, El Ouadrani K, Petitprez A. Targeting EGFR and VEGF(R) pathway cross-talk in tumor survival and angiogenesis. Pharmacol Ther. 2011;131(1):80–90.
Majidpoor J, Mortezaee K. Angiogenesis as a hallmark of solid tumors - clinical perspectives. Cell Oncol (Dordr). 2021;44(4):715–37.
Chen Z, Fadiel A, Naftolin F, Eichenbaum KD, Xia Y. Circulation DNA: biological implications for cancer metastasis and immunology. Med Hypotheses. 2005;65(5):956–61.
Holmgren L, Szeles A, Rajnavölgyi E, Folkman J, Klein G, Ernberg I, et al. Horizontal transfer of DNA by the uptake of apoptotic bodies. Blood. 1999;93(11):3956–63.
Jahr S, Hentze H, Englisch S, Hardt D, Fackelmayer FO, Hesch RD, et al. DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res. 2001;61(4):1659–65.
Choi JJ, Reich CF, Pisetsky DS. The role of macrophages in the in vitro generation of extracellular DNA from apoptotic and necrotic cells. Immunology. 2005;115(1):55–62.
Leon SA, Shapiro B, Sklaroff DM, Yaros MJ. Free DNA in the serum of cancer patients and the effect of therapy. Cancer Res. 1977;37(3):646–50.
Thierry AR, El Messaoudi S, Gahan PB, Anker P, Stroun M. Origins, structures, and functions of circulating DNA in oncology. Cancer Metastasis Rev. 2016;35(3):347–76.
Bendich A, Wilczok T, Borenfreund E. Circulating DNA as a possible factor in oncogenesis. Science. 1965;148(3668):374–6.
García-Olmo D, García-Olmo DC. Functionality of circulating DNA: the hypothesis of genometastasis. Ann N Y Acad Sci. 2001;945:265–75.
García-Olmo DC, Domínguez C, García-Arranz M, Anker P, Stroun M, García-Verdugo JM, et al. Cell-free nucleic acids circulating in the plasma of colorectal cancer patients induce the oncogenic transformation of susceptible cultured cells. Cancer Res. 2010;70(2):560–7.
García-Olmo D, García-Olmo DC, Domínguez-Berzosa C, Guadalajara H, Vega L, García-Arranz M. Oncogenic transformation induced by cell-free nucleic acids circulating in plasma (genometastasis) remains after the surgical resection of the primary tumor: a pilot study. Expert Opin Biol Ther. 2012;12(Suppl 1):S61-68.
Ichiro IS, Guarente L. NAD and sirtuins in aging and disease. Trends Cell Biol. 2014;24(8):464–71.
Covarrubias AJ, Perrone R, Grozio A, Verdin E. NAD+ metabolism and its roles in cellular processes during ageing. Nat Rev Mol Cell Biol. 2021;22(2):119–41.
Ruszkiewicz JA, Bürkle A, Mangerich A. Fueling genome maintenance: on the versatile roles of NAD+ in preserving DNA integrity. J Biol Chem. 2022;298(6): 102037.
Kirkland JL, Tchkonia T. Senolytic drugs: from discovery to translation. J Intern Med. 2020;288(5):518–36.
Baker DJ, Wijshake T, Tchkonia T, LeBrasseur NK, Childs BG, van de Sluis B, et al. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature. 2011;479(7372):232–6.
Ji S, Xiong M, Chen H, Liu Y, Zhou L, Hong Y, et al. Cellular rejuvenation: molecular mechanisms and potential therapeutic interventions for diseases. Signal Transduct Target Ther. 2023;8(1):116.
Wiggs AG, Chandler JK, Aktas A, Sumner SJ, Stewart DA. The effects of diet and exercise on endogenous estrogens and subsequent breast cancer risk in postmenopausal women. Front Endocrinol. 2021;20(12): 732255.
Collaborative Group on Hormonal Factors in Breast Cancer. Menarche, menopause, and breast cancer risk: individual participant meta-analysis, including 118 964 women with breast cancer from 117 epidemiological studies. Lancet Oncol. 2012;13(11):1141–51.
Xie Y, Xie F, Zhou X, Zhang L, Yang B, Huang J, et al. Microbiota in tumors: from understanding to application. Adv Sci (Weinh). 2022;9(21): e2200470.
Nejman D, Livyatan I, Fuks G, Gavert N, Zwang Y, Geller LT, et al. The human tumor microbiome is composed of tumor type-specific intracellular bacteria. Science. 2020;368(6494):973–80.
Gohil SH, Iorgulescu JB, Braun DA, Keskin DB, Livak KJ. Applying high-dimensional single-cell technologies to the analysis of cancer immunotherapy. Nat Rev Clin Oncol. 2021;18(4):244–56.
Aredo JV, Purington N, Su L, Luo SJ, Diao N, Christiani DC, et al. Metabolomic profiling for second primary lung cancer: a pilot case-control study. Lung Cancer. 2021;155:61–7.
Wikoff WR, Grapov D, Fahrmann JF, DeFelice B, Rom WN, Pass HI, et al. Metabolomic markers of altered nucleotide metabolism in early stage adenocarcinoma. Cancer Prev Res (Phila). 2015;8(5):410–8.
Li Y, Wang Y, Wu P. 5’-Methylthioadenosine and cancer: old molecules, new understanding. J Cancer. 2019;10(4):927–36.
Zuo W, Bai W, Gan X, Xu F, Wen G, Zhang W. Detection of lung cancer by analysis of exhaled gas utilizing extractive electrospray ionization-mass spectroscopy. J Biomed Nanotechnol. 2019;15(4):633–46.
Lindahl A, Heuchel R, Forshed J, Lehtiö J, Löhr M, Nordström A. Discrimination of pancreatic cancer and pancreatitis by LC-MS metabolomics. Metabolomics. 2017;13(5):61.
Puchades-Carrasco L, Pineda-Lucena A. Metabolomics applications in precision medicine: an oncological perspective. Curr Top Med Chem. 2017;17(24):2740–51.
Acknowledgements
Not applicable.
Funding
This work was supported by grants from the Natural Science Foundation of Guangdong Province (2021A1515012593), the National Natural Science Foundation of China (82172811, 82172750, 82260546, 82372883 and 82373129), the Guangdong Basic and Applied Basic Research Foundation (Guangdong–Guangzhou Joint Funds) (2022A1515111212), the Science and Technology Program of Guangzhou (No.2023A04J0442, No.2023A04J1257), and the Hunan Youth Science and Technology Talent Project (NO.2023RC3074).
Author information
Authors and Affiliations
Contributions
M.Y Zeng: Writing- Original draft preparation, Writing- Reviewing and Editing. A.Q Lin: Writing- Reviewing and Editing, Supervision. A.M Jiang: Supervision, Z.G Qiu: Supervision, H.M Zhang: Supervision, S.F Chen: Supervision, M.Y Xu: Supervision, Z.Q Liu: Supervision. Q Cheng: Supervision. J Zhang: Supervision. P Luo: Conceptualization, Writing- Reviewing and Editing, Supervision.
Corresponding authors
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Additional information
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Zeng, M., Lin, A., Jiang, A. et al. Decoding the mechanisms behind second primary cancers. J Transl Med 23, 115 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-025-06151-9
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-025-06151-9