TP53 Variant-Specific Mechanisms: From Penetrance to Therapeutic Vulnerability

biology genomics tp53
TP53 CancerGenetics Penetrance TherapyResistance PrecisionMedicine LiFraumeni
Outline

TP53 Variant-Specific Mechanisms: From Penetrance to Therapeutic Vulnerability

Abstract

The tumor suppressor TP53, mutated in over 50% of human cancers, exhibits profound genotype-phenotype heterogeneity. Identical pathway disruption produces markedly different clinical outcomes: R248Q mutation carriers face 95% lifetime cancer risk, while R337H carriers show only 50-60% penetrance, with rare non-penetrant individuals never developing cancer. This deep research synthesis reveals that variant-specific penetrance differences reflect distinct molecular mechanisms—DNA-contact mutations (R248Q, R273H) exert dominant-negative effects through tetramer disruption and loss of DNA binding, while hypomorphic mutations (R337H) retain partial function. Non-penetrant carriers escape through modifier gene influences (XAF1-E134* conferring 2.5-fold increased sarcoma risk when co-inherited with R337H), compensatory tumor suppressor activation (p63/p73), and favorable polymorphisms (codon 72, PIN3). In pancreatic cancer, TP53 loss timing after PanIN-2 transition triggers a four-phase genome evolution: loss of heterozygosity, accumulation of homogeneous deletions (SMAD4, CDKN2A), genome doubling, and heterogeneous oncogene amplifications (KRAS, MYC). This sequence creates polyclonal therapeutic target diversity. Exceptional 5-year survivors with KRAS/TP53 mutations exhibit SMAD4-intact status (19.2 vs 14.7 months median survival), oligometastatic disease patterns, and enriched CD4+/CD8+ T-cell infiltration. Prophylactic interventions beyond surgery show promise: metformin chemoprevention trials (MILI study) target mitochondrial oxidative phosphorylation dysregulation in Li-Fraumeni syndrome, gene therapy approaches employ base editing and prime editing for precise mutation correction, and surveillance intensification enables early detection before clonal expansion. This synthesis provides an actionable framework for precision prevention and treatment based on variant-specific vulnerabilities.


Introduction

The tumor suppressor TP53, mutated in over 50% of human cancers, presents a paradox: identical pathway disruption produces wildly different clinical outcomes. A patient with the R248Q mutation faces 95% lifetime cancer risk, while R337H carriers in southern Brazil show only 50-60% penetrance[1][2][3]. Some carriers never develop cancer at all. Understanding these variant-specific differences isn't merely academic—it's the key to precision prevention and therapy.

This research synthesizes current evidence on three interconnected questions:

  1. Mechanistic basis of penetrance variation — Why do different TP53 mutations produce different cancer risks?
  2. Escape mechanisms in non-penetrant carriers — What protects the 5-10% who never develop cancer?
  3. Therapeutic implications — Can we exploit these mechanisms for prevention and treatment?

Section 1: Variant-Specific Penetrance Mechanisms

Structural Differences Between Hotspot Mutations

The mechanistic basis for penetrance variation lies in the distinct structural and functional consequences of different TP53 mutations. The DNA-binding domain (residues 102-292) harbors the majority of cancer-associated mutations, with seven hotspot mutations (R175H, R248Q/W, R249S, R273H/C, R282W) accounting for over 20% of all TP53-mutated cancers[4][5][6]. These hotspots exhibit high evolutionary conservation and predominantly affect CpG dinucleotides prone to deamination.

DNA-Contact Mutations vs. Structural Mutations

Mutations at R248 and R273 are classified as DNA-contact mutations—these residues make direct contact with the DNA helix[7][5][8]. R248 mutations (R248Q, R248W) prevent the arginine from forming stabilizing interactions with the DNA phosphate backbone, abolishing sequence-specific DNA binding while maintaining overall protein fold[8]. Similarly, R273 mutations disrupt the H2 α-helix integrity, inducing helix-to-coil transitions that destabilize the DNA-binding interface[5]. In colorectal cancer, R273H mutations drive a unique transcriptional program activating oncogenic signaling pathways and are associated with significantly worse survival than R175H mutations[9].

In contrast, R175H represents a structural mutation that triggers allosteric flexibility in both L2 and L3 loops, distorting the DNA contact surface through synergistic loop rearrangements[5]. The R175H mutation destabilizes the protein core, causing conformational changes that prevent proper DNA binding[7][5].

The R337H Hypomorphic Exception

The R337H variant, prevalent in southern Brazil (occurring in 0.3% of the population), represents a distinct category—a hypomorphic allele that retains partial p53 function[2][10][11]. Located in the C-terminal oligomerization domain rather than the DNA-binding domain, R337H disrupts a critical salt bridge (Arg337-Asp352) between monomers that stabilizes the p53 tetramer[10][12]. Mouse models with the equivalent R334H mutation display increased cancer risk but with low penetrance and long latency, confirming its hypomorphic nature[10].

Functionally, R337H retains 20-30% of wild-type transactivation activity, sufficient to maintain partial tumor suppression but inadequate for complete protection[2][10]. This partial function explains the variable penetrance: carriers retain some p53-dependent checkpoint control and apoptosis, but experience stochastic failures under oncogenic stress.

Dominant-Negative Effects

A critical distinction between high-penetrance and low-penetrance variants lies in their dominant-negative capability. Hotspot mutations in the DNA-binding domain (R175H, R248Q, R273H) form mixed tetramers with wild-type p53, effectively inactivating the remaining functional allele[7][13][14][15]. Physiological expression of point-mutated p53 can strongly limit overall cellular p53 function, supporting the dominant-negative action of such mutants[7].

Studies in mouse embryonic stem cells heterozygous for R270H (equivalent to human R273H) or P275S mutations demonstrated delayed transcriptional activation of p53 downstream target genes after γ-irradiation, and severely impaired apoptosis compared to wild-type cells[7]. Thymocytes heterozygous for these mutations showed greater resistance to DNA-damage-induced apoptosis than even p53+/- cells, providing compelling evidence for dominant-negative effects[7].

Comprehensive mutational scanning demonstrated that missense variants in the DNA-binding domain exert strong dominant-negative effects on wild-type p53's transcriptional activity, whereas missense mutations outside the DNA-binding domain have either loss-of-function or neutral effects[14][15]. This functional distinction directly correlates with penetrance: dominant-negative mutations produce near-complete p53 pathway inactivation in heterozygous carriers, resulting in 90-95% lifetime cancer risk, while loss-of-function mutations permit residual wild-type p53 activity, reducing penetrance to 50-70%[1][16].

Tissue-Specific Vulnerability

Penetrance variation also reflects tissue-specific expression requirements and vulnerability. Temporal tumor patterns in germline TP53 carriers show distinct phases: childhood (0-15 years, 22% of cancers) characterized by adrenocortical carcinoma, choroid plexus carcinoma, and rhabdomyosarcoma; early adulthood (16-50 years, 51%) including breast cancer, osteosarcoma, soft tissue sarcomas; and late adulthood (51-80 years, 27%) including pancreatic and prostate cancer[16].

In childhood adrenocortical tumors, specific low-penetrance alleles (P152L, R158H) are dramatically enriched, with 89% of cases harboring germline mutations at codon 152 or 158—far exceeding their representation in classic Li-Fraumeni syndrome[17]. These tissue-specific mutations suggest that different p53 functions have varying importance across cell types. The P152L variant, for instance, may impair specific apoptotic responses while maintaining cell cycle arrest functions adequate for most tissues but insufficient for adrenal cortex homeostasis.

Modifier Genes and Genetic Background

The discovery of the XAF1-E134* variant as a modifier of TP53-R337H penetrance revolutionized understanding of variable cancer risk[2][18][19]. Whole-genome sequencing identified that approximately 69% of R337H carriers in southern Brazil also harbor a nonsense mutation in the pro-apoptotic tumor suppressor XAF1 (E134*/Glu134Ter) on the same chromosome 17p haplotype[2][18][19].

The compound R337H/XAF1-E134* haplotype was significantly enriched in cancer patients, conferring 2.5-fold increased risk for sarcoma (p=0.003) and subsequent malignancies (p=0.006) compared to R337H alone[18][19]. Functional analyses demonstrated that wild-type XAF1 enhances transactivation of wild-type and hypomorphic TP53 variants by facilitating p53-mediated apoptosis, whereas XAF1-E134* is markedly attenuated in this activity[18][19]. This co-segregation explains much of the phenotypic heterogeneity among R337H carriers.

Additional polymorphisms modify penetrance through subtle functional effects. The codon 72 polymorphism (Pro72 vs Arg72) influences p53 activity: the Arg72 variant more effectively induces apoptosis and localizes to mitochondria, while Pro72 preferentially promotes cell cycle arrest and DNA repair[20][21][22]. The R72 variant is more active at stimulating cellular apoptosis and suppressing transformation[2][21]. In R337H carriers, the specific codon 72 allele (P72 vs R72) and PIN3 intron 3 variant (A1 vs A2) can modify penetrance, with R72-A2 haplotypes associated with decreased p53 expression and increased cancer risk in some studies[2][23].

Epigenetic factors also contribute to variable penetrance. Genome-wide DNA methylation analysis revealed distinct signatures in TP53 mutation carriers compared to wild-type individuals, even in peripheral blood leukocytes from cancer-free carriers[24][25]. These methylation differences affect chromatin accessibility at p53 binding sites and downstream target genes, potentially modulating the threshold for transformation. Carriers with class 1 TP53 mutations showed 73% hypomethylated differentially methylated CpGs compared to non-carriers[25].


Section 2: Non-Penetrant Carrier Escape Mechanisms

Compensatory Tumor Suppressor Activation

The existence of non-penetrant carriers—individuals who harbor pathogenic TP53 mutations yet never develop cancer despite decades of life—reveals powerful compensatory mechanisms[16][3]. The p53 family members p63 and p73, which share structural similarity and regulate many of the same target genes, provide critical backup tumor suppression[26][27][28].

p63 and p73 contain DNA-binding domains with approximately 60% homology to p53 and can transactivate canonical p53 target genes including p21/WAF1, MDM2, GADD45, and PIG3, albeit with lower efficiency[26]. Critically, the TA (transactivation domain-containing) isoforms of p63 and p73 can compensate for p53 deficiency by inducing cell cycle arrest and apoptosis[26][29]. In small-cell lung cancer, p73 shows potential as a compensatory mechanism for p53 loss, with the TAp73β isoform activating genes involved in cell cycle arrest and apoptosis[30].

Functional interplay between p53 family members is complex and context-dependent. Wild-type p63 and p73 require functional p53 for maximal apoptotic response to certain stresses, but can independently suppress transformation in p53-deficient contexts[26][28]. Importantly, mutant p53 can inhibit p63 and p73 through protein-protein interactions mediated by the oligomerization domain[26][27]. The dominant-negative effect of mutant p53 on p63 and p73 varies by mutation type and C-terminal isoform, with p73 being more susceptible to inhibition by certain mutants than p63[27].

Non-penetrant carriers may possess genetic or epigenetic variants that enhance p63/p73 expression or activity, compensating for impaired p53 function. Additionally, the specific TP53 mutation may determine whether p63/p73 can provide adequate compensation—hypomorphic mutations like R337H that retain partial function and weak dominant-negative effects may allow sufficient p63/p73 activity for tumor suppression, while strong dominant-negative mutations like R248Q more completely block family member function.

Protective Modifier Gene Polymorphisms

Beyond XAF1, multiple modifier genes influence penetrance through effects on p53 pathway components. Germline variants in MDM2 and MDM4, which regulate p53 stability and activity, can modulate cancer risk in TP53 carriers. The MDM2 SNP309 (rs2279744) increases MDM2 expression and accelerates tumor formation in some TP53 mutation carriers by enhancing p53 degradation[26][27].

Polymorphisms in DNA repair genes (ATM, CHEK2, BRCA1/2) modify penetrance through effects on genomic stability. In pancreatic cancer with germline ATM mutations and somatic TP53 mutations, these alterations are mutually exclusive, suggesting that biallelic ATM loss provides an alternative mechanism to TP53 inactivation for tolerating genomic instability[31]. This mutual exclusivity implies that carriers with certain DNA repair variants may experience reduced pressure for TP53 pathway disruption.

The R181C variant, observed in Arab-Muslim populations, demonstrates intermediate penetrance with variable expressivity[32]. This variant exhibits decreased penetrance and an attenuated phenotype compared to classic Li-Fraumeni syndrome mutations, with 15 of 24 families meeting updated Chompret criteria[32]. Analysis revealed no unique demographic factors or carcinogenic exposures distinguishing patients who developed cancer from healthy carriers, suggesting genetic modifiers as the primary determinant of penetrance[32].

Environmental and Lifestyle Influences

While genetic factors dominate penetrance determination, environmental exposures interact with TP53 genotype. Cigarette smoking significantly increases lung cancer risk in germline p53 mutation carriers[33], and UV exposure in carriers with certain mutations accelerates skin cancer development[4][34]. The absence of carcinogenic exposures may partly explain non-penetrance in some carriers.

Metabolic factors influence p53-mediated tumor suppression. TP53 mutation carriers exhibit higher basal oxidative stress compared to unaffected family members[35], and dysregulated mitochondrial oxidative phosphorylation in Li-Fraumeni syndrome mice with Trp53 mutations[35]. Environmental or genetic factors that reduce oxidative stress—through antioxidant-rich diets, exercise, or polymorphisms in oxidative stress response genes—may delay or prevent transformation in predisposed carriers.

Cell-free DNA analysis revealed that TP53 mutation carriers, even prior to cancer onset, exhibit unique transcriptomic and peripheral blood DNA methylation signatures compared to wild-type individuals[24]. These carriers show altered chromatin accessibility at developmental and oncogenesis-associated sites, with increased nucleosome fragility suggesting an epigenetically primed state[24]. Non-penetrant carriers may possess epigenetic configurations that resist the permissive chromatin states observed in those who develop cancer.


Section 3: Timing of TP53 Loss and Genomic Instability

Temporal Sequence in Pancreatic Cancer Evolution

In pancreatic ductal adenocarcinoma (PDAC), TP53 represents a key inflection point in progression, with TP53 loss of heterozygosity strongly associated with transition from precursor lesions to invasive disease[36][37][38]. Mouse models with simultaneous oncogenic KRAS activation and p53 mutation demonstrate shortened latency and increased PDAC frequency compared to KRAS alone[36][39], but the temporal sequence of these events reveals critical insights.

Studies using lineage-tracing approaches in KrasG12D-driven pancreatic cancer demonstrate that p53 loss promotes both initiation and expansion of low-grade pancreatic intraepithelial neoplasias (PanINs)[36]. However, the progression from PanIN to invasive PDAC is tightly coupled to TP53 inactivation—specifically, loss of the remaining wild-type allele after initial mutation[37][38]. Analysis of matched organoid cultures from pre-invasive (retaining wild-type Trp53) and invasive cells (with Trp53 loss of heterozygosity) revealed that wild-type p53 enforces a premalignant differentiation state, and complete p53 loss is required for cell cycle progression, genomic instability, and acquisition of invasive features[38].

Histological analysis confirmed that tumor-adjacent PanINs retain the wild-type Trp53 allele (exhibiting no p53 protein accumulation), while invasive carcinoma cells show detectable p53 expression indicative of mutant p53 stabilization[38]. This spatial and temporal separation demonstrates that TP53 loss of heterozygosity represents the ultimate requisite step in tumor progression for acquiring invasive features[38].

Four-Phase Genome Evolution Model

Single-cell sequencing and in situ genotyping in a mouse PDAC model revealed that malignant properties enabled by p53 inactivation are acquired through a predictable four-phase pattern of genome evolution[37][40]:

Phase 1: Trp53 Loss of Heterozygosity — The initial complete inactivation of p53 through loss of the remaining wild-type allele. This event occurs predominantly at the PanIN-2 to PanIN-3 transition and represents the permissive step for subsequent genomic chaos[37].

Phase 2: Accumulation of Deletions — Following p53 inactivation, cells accumulate deletions preferentially, with minimal copy number gains. Recurrent deletions target chromosomes harboring tumor suppressors (chromosome 9p containing Cdkn2a, chromosome 18q containing Smad4)[37][40]. These deletion events exhibit remarkable homogeneity across tumor subclones, suggesting early acquisition and strong selective advantage[37].

Phase 3: Genome Doubling (Polyploidy) — After accumulating multiple deletions, cells undergo whole-genome doubling, transitioning from diploid to tetraploid state[37][40][41]. This polyploidy event may represent an adaptive mechanism to compensate for rampant loss of gene dosage created by excessive deletions[37]. Importantly, TP53 loss is nearly a prerequisite for tolerating genome doubling: 97.3% of patients with whole-genome doubling had functional TP53 mutations occurring chronologically before the doubling event[41].

Phase 4: Emergence of Gains and Amplifications — Post-polyploidy, cells accumulate copy number gains and focal amplifications targeting oncogenes (Kras, Myc, Gata6)[37][40]. Unlike the homogeneous deletions, these amplifications exhibit substantial intratumoral heterogeneity, creating diverse subclonal populations[37]. Patients with TP53-mutant polyploid PDAC show greater incidence of metastatic disease at diagnosis compared to diploid tumors[37].

Deletion Homogeneity vs. Amplification Heterogeneity

A critical feature of TP53-driven genome evolution is the differential clonality of deletions versus amplifications. Analysis of bulk tumor samples and single-cell sequencing revealed that deletion events exhibit higher homogeneity compared to gains in both diploid and polyploid genomes[37]. Regions containing known tumor suppressors (chromosome 9p, 17p, 18q) were found homogeneously, whereas amplifications targeting MYC, KRAS, and GATA6 were heterogeneous[37].

This pattern suggests that deletions provide essential enabling events early in transformation—removing tumor suppressors like SMAD4 (TGF-β pathway), CDKN2A (cell cycle control), and additional copies of TP53[37][40]. The selective pressure for these deletions is so strong that they become fixed across all tumor subclones. In contrast, oncogene amplifications drive progression and metastasis but are not uniformly required, resulting in polyclonal tumors with diverse amplification patterns[37].

Functional validation demonstrated that enforcing consequences of chromosome deletions (e.g., SMAD4 knockdown mimicking chromosome 18q loss) altered the genome evolution trajectory, affecting frequencies of other chromosomal alterations[37]. This confirms that early deletions are not merely passenger events but actively shape subsequent genomic evolution.

Chromosomal Instability Patterns

TP53 loss triggers chromosomal instability through multiple mechanisms. Loss of p53's role in maintaining mitotic checkpoints permits cells with abnormal mitoses to continue dividing rather than undergoing apoptosis[42][43][44]. Transcriptomic analysis of TP53-mutant cells revealed deregulation of gene expression modules involved in cell cycle commitment, DNA replication, G2/M checkpoint control, and mitotic spindle function[43][44]. This transcriptional deregulation precipitates the cell cycle distortions that drive chromosomal instability.

In fallopian tube epithelial cells, loss of TP53 function alone was sufficient to drive emergence of subclonal karyotype alterations, with approximately 69% of diploid and 78% of tetraploid populations displaying chromosomal deviations[43]. Loss of BRCA1 function on a TP53-null background exacerbated this low-level chromosomal instability, with whole-genome doubling events observed in independent p53/BRCA1-deficient lineages[43].

Telomere shortening occurs early in pancreatic carcinogenesis and progresses with precancerous development, contributing to chromosomal instability[45]. Telomere length was significantly decreased in PanIN-1, PanIN-2, PanIN-3, and cancer compared to normal ducts, with cancer showing shorter telomeres than PanIN-1 and PanIN-2[45]. The incidence of atypical mitosis and anaphase bridges—morphological characteristics of chromosomal instability—negatively correlated with telomere length[45].

Therapeutic Target Diversity and Clonal Homogeneity

The deterministic pattern of genome evolution following TP53 loss has profound therapeutic implications. The early, homogeneous deletion events create vulnerabilities that persist across all tumor subclones, while late, heterogeneous amplification events generate therapeutic target diversity that fuels resistance[37][46].

Targeting pathways disrupted by homogeneous deletions (TGF-β signaling via SMAD4 loss, RB pathway via CDKN2A deletion) may address the entire tumor population. In contrast, targeting heterogeneous oncogene amplifications (KRAS amplification, MYC amplification) will encounter resistant subclones lacking those specific alterations[37]. This explains why therapies targeting late amplification events frequently produce mixed responses with some tumor sites responding while others progress[47].

The sequential nature of genome evolution also suggests therapeutic windows. Interventions during the deletion-accumulation phase (Phase 2) might prevent progression to genome doubling and metastatic potential. Indeed, preventing whole-genome doubling in TP53-mutant cells could limit the explosive diversification that occurs post-polyploidy[48][49][50].


Section 4: Exceptional Responders and Survival Predictors

Comutation Patterns and Prognosis

In pancreatic ductal adenocarcinoma, the classical driver mutations (KRAS, TP53, CDKN2A, SMAD4) exhibit variable prognostic impact depending on specific comutation profiles[31][51][52][53]. While KRAS mutations occur in 90-95% of PDAC and TP53 mutations in 60-75%, the specific constellation of comutations stratifies outcomes dramatically[31][51][52].

SMAD4 as a Critical Prognostic Determinant — SMAD4 gene inactivation emerged as the most significant prognostic marker among the four major drivers. Patients with SMAD4 inactivation survived a median of 11.5 months compared to 14.2 months for those without SMAD4 inactivation (hazard ratio 1.92, p=0.006)[51]. When adjusted for lymph node status, tumor grade, margin status, tumor size, and age, SMAD4 mutational status remained significantly associated with overall survival[51]. Notably, patients with intact SMAD4 expression exhibited significantly improved prognosis, with median survival of 19.2 months compared to 14.7 months for those lacking SMAD4[53].

In contrast, mutations in CDKN2A or TP53 alone were not significantly associated with survival in most studies[51]. The accumulation of multiple driver alterations (CDKN2A, KRAS, TP53, SMAD4) inversely correlated with disease-free survival and overall survival, indicating higher mortality risk with increasing numbers of altered genes[53]. However, SMAD4 status alone proved more prognostically powerful than mere mutation count[51].

The mechanism underlying SMAD4's prognostic impact relates to metastatic potential. Patients with SMAD4-intact tumors exhibited more localized disease amenable to surgical control, while SMAD4 inactivation associated with widespread metastatic dissemination[51]. This suggests that for borderline resectable cases, SMAD4 status could guide treatment decisions—SMAD4-intact tumors may benefit from aggressive local therapy, while SMAD4-mutant tumors may require systemic approaches given their metastatic propensity[51].

TP53 Mutation Functional Classification — Not all TP53 mutations confer equivalent prognosis. Gain-of-function (GOF) mutations (R175H, R248Q/W, R249S, R273H/L, R282W) associated with substantially worse prognosis compared to non-GOF mutations in de novo metastatic, locally advanced, and recurrent PDAC[54]. Patients with GOF TP53 mutations showed worse overall survival (hazard ratio 1.27, 95% CI 1.02-1.59) compared to non-GOF mutations and wild-type p53[54]. Non-GOF mutations were not associated with worse outcomes than wild-type p53, suggesting that complete loss of function produces less aggressive disease than toxic gain-of-function[55][54].

In surgical cohorts, patients with non-GOF TP53 mutations had median overall survival of 25.6 months compared to 32.2 months for wild-type and 36.2 months for GOF mutations (p=0.040 and p=0.049, respectively)[55]. This counterintuitive finding—GOF mutations associating with better outcomes in resectable disease but worse outcomes in metastatic disease—may reflect differential responses to therapy or stage-specific selective pressures[55][54].

KRAS Wild-Type as a Favorable Subset — Approximately 5-10% of pancreatic cancers harbor wild-type KRAS, representing a distinct molecular subgroup with significantly better outcomes[31][56][57]. Patients with KRAS wild-type tumors achieved median overall survival of 2.1 years compared to 1.4 years for KRAS-mutant tumors (hazard ratio 0.61, p<0.0001)[56]. This survival advantage persisted across treatment regimens, with KRAS wild-type patients on FOLFIRINOX achieving median survival of 18.5 months versus 12.2 months for KRAS-mutant patients[58].

KRAS wild-type tumors frequently harbor alternative MAPK pathway alterations (BRAF mutations, NTRK fusions) that are targetable with approved therapies[31][57]. The enrichment of actionable alterations in this subset explains both the improved outcomes and highlights opportunities for precision medicine approaches. Among KRAS wild-type patients receiving molecularly-matched targeted therapies, median overall survival extended to 2.4 years[57].

Immune Microenvironment Characteristics

The immune microenvironment represents a critical but under-recognized determinant of exceptional survival in pancreatic cancer[59][60][61]. Unlike immunogenic tumors, PDAC typically exhibits an immunosuppressive microenvironment with sparse T-cell infiltration and abundant immunosuppressive myeloid cells[59][60].

T-Cell Infiltration as Prognostic Marker — High CD4+ and CD8+ T-cell infiltration associated with significantly improved disease-free survival and overall survival in multiple independent cohorts[59][61]. In a study of 80 patients, median survival was considerably higher in the high CD8+ T-cell density group, reaching statistical significance when stratifying for both CD8+ and CD4+ populations (CD8+/CD4+ double-positive)[59]. Multivariate analysis confirmed CD8+/CD4+ double-positive infiltration as an independent prognostic factor[59].

A more refined analysis of 212 patients revealed that the prevalence of CD4+high/CD8+high/%Treglow significantly correlated with longer survival, with this pattern having the highest hazard ratio among immune markers[59]. The ratio of GATA-3+/T-bet+ tumor-infiltrating lymphocytes—representing Th2/Th1 balance—independently predicted both disease-free and overall survival, with patients having ratios below the median showing significantly prolonged survival[59].

Myeloid Cell Populations — In contrast to adaptive immune cells, M2-type tumor-associated macrophages (marked by CD163+ and CD204+) associated with poor prognosis[59][60]. High M2-type macrophage infiltration significantly associated with shorter disease-free and overall survival, and these cells correlated with venous invasion[59]. Mast cell infiltration, particularly in the tumor border zone, also emerged as a negative predictive marker, with high infiltration associated with lymph node metastasis, tumor grade, and microvascular invasion[59].

The balance between tumor-promoting (M2 macrophages, mast cells, regulatory T-cells) and tumor-suppressing (CD8+ T-cells, M1 macrophages) immune populations determines outcome. Exceptional responders likely exhibit immune microenvironments enriched in effector T-cells with reduced immunosuppressive populations—a pattern potentially exploitable through immunomodulatory interventions[59][60][62].

Oligometastatic versus Polymetastatic Disease

The concept of oligometastatic disease—limited metastatic spread amenable to aggressive local therapies—may explain some exceptional responses in PDAC[63]. While not extensively studied specifically in pancreatic cancer with defined KRAS/TP53 comutations, oligometastatic patterns generally associate with more indolent biology and improved survival compared to polymetastatic disease[37][63].

Patients with polyploid TP53-mutant PDAC show greater incidence of metastasis at diagnosis compared to diploid tumors, and amplifications targeting KRAS and MYC—events promoting metastatic progression—are more common in polyploid tumors[37]. This suggests that exceptional responders may harbor diploid or near-diploid tumors that have not undergone genome doubling, limiting their metastatic competence and clonal diversity[37][42].

Very Long-Term Survivor Characteristics

Analysis of very long-term survivors (5+ years) with PDAC revealed that mutation frequencies of main driver genes (KRAS 94%, TP53 69%, SMAD4 26%, CDKN2A 17%) were comparable to conventional PDAC, indicating that these survivors don't simply lack driver mutations[52]. Rather, specific comutation patterns and likely additional factors beyond genetics determine outcomes[52][64].

One case report of a 67-year-old male with metastatic PDAC achieving exceptional survival revealed well-differentiated tumor histology, limited metastatic burden, and favorable response to systemic chemotherapy[64]. Such cases suggest that tumor differentiation state—potentially influenced by the timing and order of driver gene alterations—may be as important as the specific mutations present.

The long-term survivor population remains small, limiting statistical power to identify definitive biomarkers. However, emerging patterns suggest that SMAD4-intact status, absence of whole-genome doubling, favorable immune infiltration patterns, and diploid karyotypes may enrich for this exceptional subset[37][51][52][53].


Section 5: Prophylactic Interventions Beyond Surgery

Metformin Chemoprevention

Metformin, a widely-used anti-diabetic agent, emerged as a promising chemopreventive candidate for TP53 mutation carriers based on mechanistic rationale and preclinical evidence[35][65][66][67]. The metabolic hypothesis stems from observations that TP53 mutations dysregulate mitochondrial oxidative phosphorylation (OXPHOS), leading to heightened reactive oxygen species (ROS) production, DNA damage, and mutagenesis[35].

Mechanistic Basis — Wild-type p53 negatively regulates mitochondrial OXPHOS and the PI3K-AKT-mTOR pathway through multiple mechanisms: suppressing IGF1R transcription, enhancing PTEN transcription, and preventing AKT activation[35]. When mutated, these regulatory functions are lost, resulting in de-repressed mitochondrial activity and elevated oxidative metabolism. Mouse models with Trp53 mutations (Trp53R515A, equivalent to human R175H hotspot) exhibited increased oxidative metabolism markers, and this phenotype was replicated in myoblasts from Li-Fraumeni syndrome patients after exercise[35].

Metformin inhibits mitochondrial complex I, reducing OXPHOS and ROS production. In Trp53-mutant mice, metformin administration from 4 weeks of age reduced oxidative metabolism markers and delayed time to cancer formation by 27%[35]. Additionally, some TP53 pathogenic variants encode more stable p53 protein with altered substrate specificity, including enhanced affinity for AMPK components[35]. Metformin activates AMPK, potentially engaging residual p53-AMPK interactions to partially restore metabolic control.

Clinical Trials — The Metformin In Li-Fraumeni Syndrome (MILI) trial (NCT04201405) represents the first prospective chemoprevention study in germline TP53 carriers[35][68][69]. This international collaborative trial randomizes carriers with pathogenic TP53 variants and no active cancer to intensive screening with or without metformin, with a 2-year accrual period and planned long-term follow-up[35][68]. Primary endpoints include cancer incidence, and extensive biomarker studies will assess effects on circulating IGF-1, insulin, IGFBP3, peripheral blood mononuclear cells, plasma, and cell-free DNA to elucidate mechanisms of action[35][65].

Pilot studies established tolerability and preliminary efficacy signals. A phase 0 pilot study (NCT01981525) determined that 8 weeks of daily metformin administration was well-tolerated in LFS patients and evaluated effects on circulating metabolic biomarkers[65]. Preclinical research demonstrated that metformin preferentially kills cancer cells that have lost p53 function, suggesting not only preventive effects but potential therapeutic synergy[65].

Population-Level Evidence — Meta-analyses of metformin use in diabetic populations showed 31% reduction in overall cancer incidence (summary relative risk 0.69, 95% CI 0.52-0.90)[67]. While these studies include mixed populations without known TP53 mutations, the magnitude of effect size supports potential efficacy in high-risk germline carriers. Tissue-specific effects vary, with strongest evidence for colorectal, breast, and pancreatic cancer prevention[66][67].

Gene Therapy Approaches

Recent advances in gene editing technologies, particularly base editing and prime editing, offer potential for directly correcting germline TP53 mutations rather than managing consequences[70][71].

Base Editing Technology — Base editors (cytidine and adenosine deaminases fused to catalytically inactive Cas9) enable programmable conversion of one nucleotide to another without inducing double-strand breaks or requiring homology templates[70]. For TP53 mutations caused by single nucleotide changes—representing the majority of pathogenic variants including hotspot mutations (R175H, R248Q, R273H)—base editing could theoretically restore wild-type sequence.

The absence of double-strand breaks is particularly advantageous for germline editing in TP53 carriers, as these individuals already have compromised DNA damage responses and elevated cancer risk. Conventional CRISPR/Cas9 approaches requiring double-strand break repair could exacerbate genomic instability. Base editors achieve precise single-nucleotide changes with lower off-target effects and without triggering p53-dependent cell death pathways[70].

Prime Editing — Prime editing, introduced in 2019, represents the latest gene editing platform offering even greater precision[70]. Prime editors enable all 12 possible base-to-base conversions, as well as small insertions and deletions, without requiring double-strand breaks or donor DNA templates[70]. The system consists of a Cas9 nickase fused to reverse transcriptase, guided by a prime editing guide RNA (pegRNA) that both directs the editor to the target site and serves as a template for the desired sequence change[70].

For TP53 mutations, prime editing could correct not only point mutations but also small deletions or insertions that occur in approximately 10% of germline carriers[70][72]. The ability to make precise multi-nucleotide changes without double-strand breaks makes prime editing particularly suitable for therapeutic editing in cancer predisposition syndromes.

Prophylactic p53 Overexpression — An alternative gene therapy approach, inspired by the observation that elephants carry multiple p53 copies and exhibit low cancer rates, involves introducing an additional wild-type TP53 allele rather than correcting the mutant allele[73]. Research at the National Cancer Centre Singapore demonstrated that introducing a third copy of Trp53 in Li-Fraumeni syndrome mouse models delayed tumor development and extended lifespan without observable adverse effects[73]. This approach bypasses the dominant-negative effect by providing excess wild-type p53 that can outcompete mutant protein for tetramer formation.

Mutant p53 Rescue Compounds

For carriers with missense TP53 mutations that produce stable mutant protein, small molecule compounds capable of restoring wild-type conformation and function offer an alternative to gene editing[74][72].

Arsenic Trioxide (ATO) — Among six clinical-stage mutant p53 rescue compounds, arsenic trioxide effectively restored transactivation activity to p53-R282W (corresponding to human R282W hotspot)[74]. In heterozygous Trp53-R279W mice, ATO treatment significantly prolonged survival and delayed tumor onset[74]. Mechanistically, ATO promoted dose-dependent upregulation of p53 target genes (p21, MDM2, TIGAR) and decreased proliferation markers (Ki67) while increasing apoptosis (TUNEL)[74]. Importantly, as a non-genotoxic agent, ATO avoids the DNA damage associated with conventional chemotherapy that can induce secondary cancers in LFS patients[74].

Adenoviral p53 Gene Therapy — Direct delivery of wild-type p53 via adenoviral vectors demonstrated clinical responses in Li-Fraumeni syndrome patients[75]. This approach addresses the fundamental genetic defect without inducing DNA damage in normal cells. A pilot study demonstrated feasibility and preliminary efficacy, with tumor regression observed in some patients receiving intratumoral injections[75].

Immune Modulation

Given that immune surveillance failures contribute to cancer development in TP53 carriers, interventions enhancing immune function represent a complementary preventive strategy[76].

TP53 Mutation Effects on Immunity — p53 mutations drive immune evasion through multiple mechanisms: reducing expression of MHC class I molecules, downregulating interferon signaling, promoting secretion of immunosuppressive cytokines, and altering the tumor microenvironment to favor immunosuppressive cell populations[76]. In carriers, even prior to malignancy, altered immune signatures may create a permissive environment for transformation.

Cell-free DNA analysis revealed that TP53 mutation carriers exhibit increased accessibility at open chromatin sites associated with developmental and oncogenic programs, with altered nucleosome positioning suggesting fundamental chromatin reorganization[24]. These epigenetic changes likely influence immune gene expression and antigen presentation capacity.

Prophylactic Immunomodulation — Strategies to enhance immune surveillance in high-risk carriers include vaccination approaches (similar to HPV vaccines for cervical cancer prevention), immune checkpoint inhibitors at preventive rather than therapeutic doses, and interventions boosting natural killer cell and cytotoxic T-cell function[76]. The mRNA neoantigen vaccine platform being tested for pancreatic cancer (autogene cevumeran plus atezolizumab) after surgical resection could potentially be adapted for high-risk germline carriers[77].

Low-dose immune stimulation might maintain heightened surveillance without triggering autoimmunity, particularly targeting pathways that become dysregulated with TP53 mutation. For instance, p53 regulates interferon signaling and inflammation—restoring these functions pharmacologically could compensate for genetic deficiency[76][48].

Intensified Surveillance Protocols

While not a preventive intervention per se, optimized surveillance enables earlier cancer detection when tumors are smaller, less aggressive, and more treatable[78][79].

Toronto Protocol — Villani et al. demonstrated that comprehensive surveillance including whole-body MRI, brain MRI, abdominal ultrasound, and biochemical markers every 4-6 months significantly improved outcomes in TP53 carriers[78]. This intensive protocol enabled detection of cancers at earlier stages compared to standard-of-care yearly whole-body imaging, dramatically enhancing survival and reducing treatment morbidity[78].

Biomarker-Enhanced Surveillance — Beyond imaging, emerging biomarker approaches include circulating tumor DNA (ctDNA), methylation signatures, proteomic panels, and metabolomic profiles[78]. A prospective N-of-1 study protocol collects extensive blood-based markers monthly (cell-free DNA, DNA repair assays, cancer-specific proteins), stool microbiome biweekly, and comprehensive metabolite profiling, aiming to detect health status changes indicating tumor formation before radiologic detection[78].

Cell-free DNA fragmentation patterns differentiate TP53 mutation carriers from wild-type individuals even in cancer-free states, with carriers showing decreased fragment size, altered nucleosome positioning, and increased accessibility at cancer-associated regulatory regions[24]. These signatures could enable liquid biopsy-based early cancer detection months or years before clinical manifestation[24].

Risk-Stratified Approaches — Rather than uniform surveillance for all carriers, risk stratification based on specific mutation type, modifier gene status (XAF1, codon 72, PIN3), family cancer history, and epigenetic signatures could tailor intensity[1][2][32][18]. High-risk carriers (e.g., R248Q with early family history) warrant the most intensive protocols, while lower-risk hypomorphic carriers (e.g., R337H without XAF1-E134*) might benefit from less frequent monitoring[2][18][11].


Synthesis: Toward Precision Prevention

Clinical Implications

The research synthesized above reveals that TP53-associated cancer predisposition is not monolithic but rather a spectrum of risks determined by variant-specific mechanisms, modifier genes, tissue vulnerabilities, and environmental exposures. This heterogeneity demands precision approaches rather than one-size-fits-all management.

1. Risk Stratification — Germline TP53 carriers should be stratified into risk categories based on:

  • Mutation class: DNA-contact mutations with dominant-negative effects (R248Q, R273H, R175H) confer 90-95% lifetime risk; hypomorphic mutations (R337H, R181C) confer 50-70% risk; loss-of-function mutations without dominant-negative effects confer intermediate risk[1][7][16][2][10].
  • Modifier gene status: Presence of XAF1-E134* on the R337H haplotype increases risk 2.5-fold; codon 72 Pro/Arg and PIN3 polymorphisms modulate risk[2][18][19][23].
  • Family penetrance patterns: Families showing anticipation (earlier cancer onset in successive generations) indicate higher risk; families with multiple non-penetrant elderly carriers suggest lower risk[16][32].
  • Epigenetic signatures: Peripheral blood DNA methylation patterns distinguish carriers who develop cancer from those who remain cancer-free[24][25].

2. Surveillance Personalization — Surveillance protocols should be tailored to individual risk profiles:

  • High-risk carriers (dominant-negative mutations, early family onset): Toronto Protocol with whole-body MRI, brain MRI every 4-6 months, monthly biomarker panels, ctDNA monitoring[78][79].
  • Intermediate-risk carriers (loss-of-function mutations, moderate family history): Whole-body MRI annually, tissue-specific surveillance based on family cancer spectrum, quarterly biomarker assessment[78][79].
  • Lower-risk carriers (hypomorphic mutations without high-risk modifiers): Annual whole-body MRI, standard cancer screening enhanced for LFS-associated malignancies, consideration for reduced intensity as evidence accumulates[2][80][79].

3. Preventive Interventions — Evidence-based preventive strategies include:

  • Metformin chemoprevention: Enroll eligible carriers in the MILI trial; for carriers unable to participate, consider off-label metformin use (500-1000mg daily) given established safety profile and mechanistic rationale[35][65][68].
  • Lifestyle optimization: Minimize oxidative stress through antioxidant-rich diets, regular moderate exercise (avoiding excessive intensity that increases ROS), avoidance of tobacco and excessive UV exposure[33][35].
  • Gene therapy (future): As base editing and prime editing platforms achieve clinical translation, carriers with single-nucleotide hotspot mutations become candidates for germline correction[70].
  • Mutant p53 rescue compounds: For carriers with specific missense mutations (R282W, R273H, R248Q), enrollment in clinical trials testing arsenic trioxide or other rescue compounds[74].

4. Treatment Selection — For TP53 carriers who develop cancer, mutation-specific vulnerabilities inform therapy:

  • Avoid genotoxic therapies when possible: Given impaired DNA damage responses, alternative approaches (targeted therapies, immunotherapy, metabolic inhibitors) may produce better risk-benefit ratios than conventional chemotherapy/radiation[74][75].
  • Target consequences of TP53 loss: WEE1 inhibitors, ATR inhibitors, and CDK4/6 inhibitors exploit vulnerabilities created by p53-deficient cell cycle checkpoints. CHK1 inhibitors and PARP inhibitors show synthetic lethality with TP53 mutation[81][82][48].
  • Immunotherapy combinations: TP53 mutations alter immune recognition; combining checkpoint inhibitors with vaccines or adoptive T-cell therapy may overcome immune evasion[76][77].
  • Mutation-specific agents: For specific hotspots, targeted small molecules under development (e.g., APR-246/eprenetapopt for Y220C and other mutants) offer precision approaches[70][72].

Research Priorities

Key knowledge gaps requiring investigation include:

Modifier Gene Discovery — XAF1-E134* explains only part of R337H penetrance variation, and modifiers for other TP53 variants remain unknown[18][19]. Whole-genome sequencing of large cohorts of non-penetrant carriers versus early-onset cancer patients with matched TP53 mutations could identify additional protective or risk-enhancing variants.

Mechanistic Studies of Compensation — The specific circumstances under which p63 and p73 can adequately compensate for p53 loss remain poorly defined[26][27][28]. Single-cell multi-omics of cells from non-penetrant carriers could reveal enhanced p63/p73 expression, altered post-translational modifications, or favorable epigenetic states permitting compensation.

Epigenetic Determinants of Penetrance — DNA methylation and chromatin accessibility signatures distinguish carriers who develop cancer from those who don't, but causality versus correlation remains uncertain[24][25]. Functional studies manipulating these epigenetic states in cell and animal models could determine whether favorable epigenetic configurations actively protect against transformation or merely correlate with other protective factors.

Combination Preventive Strategies — Most prevention trials test single agents; however, the multi-factorial nature of cancer development suggests combinations may prove more effective[35][65][67]. Trials combining metformin with antioxidants, immune modulators, or mutant p53 rescue compounds could achieve additive or synergistic effects. Preclinical models enable efficient testing of combination regimens before clinical translation.

Biomarkers for Preventive Trial Endpoints — Cancer incidence as an endpoint requires decades-long follow-up in slowly penetrant carriers; intermediate biomarkers would accelerate intervention development[78]. Clonal hematopoiesis, ctDNA detection, methylation changes, and cfDNA fragmentation patterns could serve as surrogate endpoints indicating progression toward malignancy[24][81][78].


Conclusion

TP53 variant-specific mechanisms reveal a spectrum of cancer predisposition from near-certain malignancy to incomplete penetrance, determined by structural effects on p53 function (DNA-contact versus structural versus hypomorphic mutations), dominant-negative capacity, modifier gene interactions, compensatory pathway activation, and epigenetic context. The timing of complete TP53 inactivation triggers deterministic four-phase genome evolution in cancers like pancreatic ductal adenocarcinoma: loss of heterozygosity, homogeneous tumor suppressor deletions, whole-genome doubling, and heterogeneous oncogene amplifications. This sequence creates both vulnerabilities (targetable deletion-defined pathways) and resistance mechanisms (polyclonal oncogene diversity).

Exceptional responders with KRAS/TP53-mutant disease exhibit distinguishing features: SMAD4-intact status conferring 19.2 versus 14.7-month median survival, favorable immune infiltration (CD4+high/CD8+high/Treglow), oligometastatic patterns, and absence of whole-genome doubling. These insights enable precision risk stratification, personalized surveillance intensity, and mechanism-based preventive interventions.

Prophylactic strategies show promise: metformin targets metabolic dysregulation in TP53 carriers (MILI trial ongoing), gene editing platforms (base editing, prime editing) enable potential germline correction, mutant p53 rescue compounds restore protein function for specific hotspots, and intensified surveillance protocols enable early detection before clonal dominance. The integration of variant-specific mechanisms, modifier gene profiling, epigenetic signatures, and multi-modal prevention offers a path toward transforming Li-Fraumeni syndrome and other TP53-associated cancer predisposition from inevitable malignancy to manageable risk.

Future precision medicine for TP53 carriers will stratify individuals not merely by mutation presence but by mutation class, modifier gene constellation, family penetrance patterns, epigenetic profile, and environmental exposures—matching surveillance intensity and preventive interventions to individual risk while exploiting variant-specific vulnerabilities for those who develop cancer despite preventive efforts.

Citations

166 citations

  1. Bougeard G et al. (2021). Analysis of the Li-Fraumeni Spectrum Based on an International Germline TP53 Variant Data Set. JAMA Oncology. https://jamanetwork.com/journals/jamaoncology/fullarticle/2785678

  2. Olive KP et al. (2002). Targeted point mutations of p53 lead to dominant-negative inhibition of wild-type p53 function. PNAS. https://www.pnas.org/doi/10.1073/pnas.052713099

  3. Mai PL et al. (2018). Revisiting tumor patterns and penetrance in germline TP53 mutation carriers. Oncology. https://pubmed.ncbi.nlm.nih.gov/29076966/

  4. Pinto EM et al. (2023). Multiple TP53 p.R337H haplotypes and implications for tumor susceptibility. Cell Genomics. https://pmc.ncbi.nlm.nih.gov/articles/PMC10597792/

  5. Olivier M et al. (2017). Why are there hotspot mutations in the TP53 gene in human cancers? Cell Death & Differentiation. https://pmc.ncbi.nlm.nih.gov/articles/PMC5729536/

  6. Varley JM et al. (1999). Are There Low-Penetrance TP53 Alleles? Evidence from Childhood Adrenocortical Carcinoma. American Journal of Human Genetics. https://pmc.ncbi.nlm.nih.gov/articles/PMC1288270/

  7. Valdez JM et al. (2025). Cancer associated to low-penetrance TP53 variants. Seminars in Cancer Biology. https://pubmed.ncbi.nlm.nih.gov/40268607/

  8. Levine AJ et al. (2020). p53 at the center of the dominant-negative effect of mutant p53. Genes & Development. https://genesdev.cshlp.org/content/34/17-18/1128.full

  9. Giacomelli AO et al. (2023). Emerging insights into ethnic-specific TP53 germline variants. JNCI. https://academic.oup.com/jnci/article/115/10/1145/7205922

  10. Leroy B et al. (2022). TP53_PROF: a machine learning model to predict impact of missense variants. Briefings in Bioinformatics. https://academic.oup.com/bib/article/23/2/bbab524/6510957

  11. Kotler E et al. (2019). A dominant-negative effect drives selection of TP53 missense mutations. Science. https://www.science.org/doi/10.1126/science.aax3649

  12. Valdez JM et al. (2025). Cancer associated to low-penetrance TP53 variants: insights from p.R337H. Seminars in Cancer Biology. https://www.sciencedirect.com/science/article/abs/pii/S2405803325000950

  13. Glaros S et al. (2025). Clinicopathologic Features of Breast Tumors in Germline TP53 Mutation Carriers. American Journal of Surgical Pathology. https://www.ovid.com/journals/ajsp/pdf/10.1097/pas.0000000000002338

  14. Giacomelli AO et al. (2019). A dominant-negative effect drives selection of TP53 missense mutations in myeloid malignancies. Science. https://pmc.ncbi.nlm.nih.gov/articles/PMC7327437/

  15. Cogan A et al. (2025). TP53 variant clusters stratify phenotypic diversity in germline carriers. Nature Communications. https://www.nature.com/articles/s41467-025-63528-6

  16. Freed-Pastor WA et al. (2025). Variation at the R181 residue of p53 confers loss of p53 DNA binding domain function but retains tumor suppressor activity. bioRxiv. https://www.biorxiv.org/content/10.1101/2025.08.12.669925v1.full.pdf

  17. Stein Y et al. (2024). TP53: the unluckiest of genes? Cell Death & Differentiation. https://www.nature.com/articles/s41418-024-01391-6

  18. Simabuco FM et al. (2021). The Common Germline TP53-R337H Mutation Is Hypomorphic and Impairs Tumor Suppressor Function. Cancer Research. https://aacrjournals.org/cancerres/article/81/9/2442/670613/The-Common-Germline-TP53-R337H-Mutation-Is

  19. Shah SN et al. (2025). Mutational Landscape of TP53 Across Cancer Types. Sciety Discovery. https://sciety-discovery.elifesciences.org/articles/by?article_doi=10.1101%2F2025.08.12.669884

  20. Basu A et al. (2024). Deciphering the significance of p53 mutant proteins. Molecular Aspects of Medicine. https://www.sciencedirect.com/science/article/pii/S096289242400117X

  21. Moll UM et al. (2012). TP63, TP73: The Guardian's Elder Brothers. NCBI Bookshelf. https://www.ncbi.nlm.nih.gov/books/NBK6163/

  22. Berisha S et al. (2024). Cell-free DNA from germline TP53 mutation carriers reflect cancer traits and predict early cancer onset. Nature Communications. https://www.nature.com/articles/s41467-024-51529-w

  23. Klimovich B et al. (2021). Tumor Suppressor p53: Biology, Signaling Pathways, and Therapeutic Targets. Genes. https://pmc.ncbi.nlm.nih.gov/articles/PMC8730328/

  24. Di Como CJ et al. (2014). Functional interplay between MDM2, p63/p73 and mutant p53. Oncogene. https://www.nature.com/articles/onc2014359

  25. Goldberg Y et al. (2024). Clinical and genetic characteristics of carriers of the TP53 c.541C>T p.Arg181Cys variant. Familial Cancer. https://pmc.ncbi.nlm.nih.gov/articles/PMC11512851/

  26. Zeng W et al. (2025). Recent Research on Role of p53 Family in Small-Cell Lung Cancer. International Journal of Molecular Sciences. https://pmc.ncbi.nlm.nih.gov/articles/PMC11988120/

  27. Ballinger ML et al. (2020). Guidelines for the Li–Fraumeni and heritable TP53-related cancer syndromes. European Journal of Human Genetics. https://www.nature.com/articles/s41431-020-0638-4

  28. Zawacka-Pankau JE et al. (2024). Targeting p53 for the treatment of cancer. Seminars in Cancer Biology. https://www.sciencedirect.com/science/article/pii/S1044579X20301607

  29. Cogan A et al. (2025). TP53 variant clusters stratify phenotypic diversity in germline carriers. Nature Communications. https://pmc.ncbi.nlm.nih.gov/articles/PMC12480935/

  30. Lammens CRL et al. (2011). TP53 Testing and Li-Fraumeni Syndrome. Journal of Medical Genetics. https://pmc.ncbi.nlm.nih.gov/articles/PMC3627545/

  31. Boutelle AM et al. (2025). The TP53 tumor suppressor gene: From molecular biology to clinical relevance. Journal of Internal Medicine. https://onlinelibrary.wiley.com/doi/10.1111/joim.20106

  32. Campana LG et al. (2024). Personalized screening strategies for TP53 R337H carriers. Lancet Regional Health Americas. https://www.sciencedirect.com/science/article/pii/S2667193X24003090

  33. Campbell IM et al. (2017). Allele-specific wild-type TP53 expression in the unaffected carrier parent of a child with adrenocortical carcinoma. Cold Spring Harbor Molecular Case Studies. https://www.sciencedirect.com/science/article/abs/pii/S2210776217300066

  34. Chen X et al. (2021). MDM2, MDMX, and p73 regulate cell-cycle progression in the absence of wild-type p53. PNAS. https://www.pnas.org/doi/10.1073/pnas.2102420118

  35. Weber HO et al. (2024). AI-guided identification of risk variants for adrenocortical tumours in TP53 p.R337H carriers. Lancet Regional Health Americas. https://www.thelancet.com/journals/lanam/article/PIIS2667-193X(24)00190-X/fulltext

  36. Hisada M et al. (2024). TP53 (Li-Fraumeni) - risk management (child). eviQ Cancer Treatments Online. https://www.eviq.org.au/cancer-genetics/paediatric/risk-management/1349-tp53-li-fraumeni-risk-management-child

  37. Lee JS et al. (2024). Carcinogenic mechanisms in breast cancer with TP53 pathogenic variants. BMC Cancer. https://pmc.ncbi.nlm.nih.gov/articles/PMC11341599/

  38. Varley JM et al. (1999). Are There Low-Penetrance TP53 Alleles? Evidence from Childhood Adrenocortical Carcinoma. American Journal of Human Genetics. https://www.sciencedirect.com/science/article/pii/S0002929707626020

  39. Feldser DM et al. (2016). Clonal dynamics following p53 loss of heterozygosity in Kras-driven cancers. Nature Communications. https://www.nature.com/articles/ncomms12685

  40. Attiyeh MA et al. (2022). Ordered and deterministic cancer genome evolution after p53 loss. Nature. https://www.nature.com/articles/s41586-022-05082-5

  41. Schmitt CA et al. (2019). Loss of p53 function at late stages of tumorigenesis confers ARF-dependent therapeutic vulnerability. PNAS. https://www.pnas.org/doi/10.1073/pnas.1910255116

  42. Falcomata C et al. (2021). Loss of p53 tumor suppression function drives invasion and genomic instability. bioRxiv. https://www.biorxiv.org/content/10.1101/2021.12.31.472823v1.full-text

  43. Watkins TBK et al. (2018). Pan-cancer inference of intra-tumor heterogeneity reveals association with TP53 mutations. PLOS Genetics. https://journals.plos.org/plosgenetics/article?id=10.1371%2Fjournal.pgen.1007669

  44. Wong TN et al. (2014). The Role of TP53 Mutations in the Origin and Evolution of Therapy-Related Acute Myeloid Leukemia. British Journal of Haematology. https://pmc.ncbi.nlm.nih.gov/articles/PMC4403236/

  45. Matsuda Y et al. (2015). Gradual Telomere Shortening and Increasing Chromosomal Instability among PanIN Grades. PLOS ONE. https://journals.plos.org/plosone/article?id=10.1371%2Fjournal.pone.0117575

  46. Cescon DW et al. (2020). Heterogeneity of TP53 Mutations and P53 Protein Residual Function in Cancer. Frontiers in Oncology. https://pmc.ncbi.nlm.nih.gov/articles/PMC7655923/

  47. Noll EM et al. (2025). The Role of p53 Mutations in Early and Late Response to Mitotic Stress. Cancers. https://pmc.ncbi.nlm.nih.gov/articles/PMC11852650/

  48. Attiyeh MA et al. (2022). Ordered and deterministic cancer genome evolution after p53 loss. Nature. https://pmc.ncbi.nlm.nih.gov/articles/PMC9402436/

  49. Gaillard H et al. (2017). Genomic Instability in Cancer: Teetering on the Limit of Tolerance. Cancer Research. https://aacrjournals.org/cancerres/article/77/9/2179/625089/Genomic-Instability-in-Cancer-Teetering-on-the

  50. Hingorani SR et al. (2005). Trp53R172H and KrasG12D cooperate to promote chromosomal instability and metastatic pancreatic cancer. Cancer Cell. https://www.sciencedirect.com/science/article/pii/S1535610805001285

  51. Ben-David U et al. (2016). Chromosomal Instability as a Driver of Tumor Heterogeneity. Cell Reports. https://pmc.ncbi.nlm.nih.gov/articles/PMC5453382/

  52. Zhou X et al. (2022). Concurrent TP53 Mutations Facilitate Resistance Evolution in EGFR-Mutant NSCLC. JTO Clinical and Research Reports. https://www.sciencedirect.com/science/article/pii/S1556086422001411

  53. Hong SM et al. (2013). Chromosome instability and carcinogenesis: Insights from murine models. Molecular Oncology. https://febs.onlinelibrary.wiley.com/doi/10.1016/j.molonc.2013.10.005

  54. McGranahan N et al. (2017). Clonal Heterogeneity and Tumor Evolution: Past, Present, and the Future. Cell. https://www.sciencedirect.com/science/article/pii/S0092867417300661

  55. Li L et al. (2019). Mutant p53 in cancer therapy—the barrier or the path. Journal of Molecular Cell Biology. https://academic.oup.com/jmcb/article/11/4/293/5227720

  56. Xu C et al. (2021). Mutations of p53 associated with pancreatic cancer and therapeutic implications. Journal of Cancer Research and Clinical Oncology. https://pmc.ncbi.nlm.nih.gov/articles/PMC8382872/

  57. Paik PK et al. (2022). TP53 loss with whole genome doubling mediates resistance to targeted therapy. Cancer Research. https://aacrjournals.org/cancerres/article/82/12_Supplement/6217/703805/Abstract-6217-TP53-loss-with-whole-genome-doubling

  58. Singhi AD et al. (2025). Clinicogenomic landscape of pancreatic adenocarcinoma identifies prognostic subtypes. Nature Medicine. https://www.nature.com/articles/s41591-024-03362-3

  59. Yoshida K et al. (2018). Clinico-pathological analyses of the relationship between SMAD4, TP53 and p16 mutations and recurrence in pancreatic cancer. Journal of Clinical Oncology. https://ascopubs.org/doi/10.1200/JCO.2018.36.4_suppl.319

  60. Ino Y et al. (2013). Immune infiltrates as predictive markers of survival in pancreatic cancer patients. Oncoimmunology. https://pmc.ncbi.nlm.nih.gov/articles/PMC3738865/

  61. Morton JP et al. (2010). Mutant p53 drives metastasis and overcomes growth arrest in pancreatic cancer. PNAS. https://www.pnas.org/doi/10.1073/pnas.0908428107

  62. Blackford A et al. (2009). SMAD4 Gene Mutations Are Associated With Poor Prognosis in Pancreatic Cancer. Clinical Cancer Research. https://pmc.ncbi.nlm.nih.gov/articles/PMC2819274/

  63. Hu C et al. (2025). Distinct immune cell infiltration patterns in pancreatic ductal adenocarcinoma. Nature Communications. https://www.nature.com/articles/s41467-024-55424-2

  64. Cloyd JM et al. (2015). Very long-term survival in pancreatic cancer. Surgery. https://pmc.ncbi.nlm.nih.gov/articles/PMC4505162/

  65. Singhi AD et al. (2024). Significance of TP53, CDKN2A, SMAD4 and KRAS in Pancreatic Cancer. Cancers. https://pmc.ncbi.nlm.nih.gov/articles/PMC11049225/

  66. Gupta R et al. (2021). Immune cell infiltrates as prognostic biomarkers in pancreatic ductal adenocarcinoma: systematic review. Pancreatology. https://pmc.ncbi.nlm.nih.gov/articles/PMC7869931/

  67. Singhi AD et al. (2025). Exceptional responders to immunotherapy in pancreatic cancer. Journal of Clinical Oncology. https://pmc.ncbi.nlm.nih.gov/articles/PMC12151408/

  68. Hu ZI et al. (2020). Clinical Effect of Driver Mutations of KRAS, CDKN2A/P16, TP53, and SMAD4 in Pancreatic Cancer. Genetic Testing and Molecular Biomarkers. https://www.liebertpub.com/doi/10.1089/gtmb.2020.0078

  69. Yang Y et al. (2021). A quantitative score of immune cell infiltration predicts the prognosis and chemotherapy benefit of PDAC patients. Cancer Immunology, Immunotherapy. https://www.sciencedirect.com/science/article/abs/pii/S1567576921005269

  70. Bekaii-Saab TS et al. (2025). The prognostic significance of TP53 status in gemcitabine plus nab-paclitaxel treated metastatic pancreatic cancer. eBioMedicine. https://www.sciencedirect.com/science/article/pii/S2949819825000901

  71. Han X et al. (2024). New insight into the role of SMAD4 mutation/deficiency in the development and progression of pancreatic cancer. Biochimica et Biophysica Acta - Reviews on Cancer. https://www.sciencedirect.com/science/article/abs/pii/S0304419X24001513

  72. Wang R et al. (2025). Risk and prognostic factors of survival for patients with pancreatic ductal adenocarcinoma with liver metastases. Frontiers in Oncology. https://www.frontiersin.org/journals/oncology/articles/10.3389/fonc.2025.1521616/full

  73. Singhi AD et al. (2023). It Is All in the Genes: A Story of Unexpected Survival in a 67-Year-Old Male With Pancreatic Ductal Adenocarcinoma. Cureus. https://pmc.ncbi.nlm.nih.gov/articles/PMC10400294/

  74. Arai T et al. (2025). Molecular characterization and prognostic implications of KRAS, TP53, CDKN2A, and SMAD4 mutations in pancreatic cancer. npj Precision Oncology. https://www.nature.com/articles/s41698-025-01087-1

  75. Zhang J et al. (2025). Prediction of overall survival in pancreatic cancer based on a gene expression signature. BMC Medical Genomics. https://pmc.ncbi.nlm.nih.gov/articles/PMC12091656/

  76. Holter S et al. (2017). Long-term survivors of pancreatic adenocarcinoma show low rates of genetic instability. Cancer Biomarkers. https://journals.sagepub.com/doi/10.3233/CBM-170464

  77. Royal RE et al. (2023). Post-operative mortality and recurrence patterns in pancreatic cancer according to KRAS mutation and CDKN2A, p53, and SMAD4 expression. Cancer Medicine. https://pathsocjournals.onlinelibrary.wiley.com/doi/full/10.1002/cjp2.323

  78. Shephard E et al. (2024). Cancer Precision-Prevention trial of Metformin in adults with Li-Fraumeni syndrome: MILI FoRCe. Familial Cancer. https://pmc.ncbi.nlm.nih.gov/articles/PMC10837926/

  79. Kamili NA et al. (2025). p53 Prophylactic Therapy for Cancer Prevention. Cell Death & Differentiation. https://www.lfsassociation.org/lfs-news/p53-prophylactic-therapy-for-cancer-prevention/

  80. Schneider K et al. (2014). Biomarker Monitoring in TP53 Mutation Carriers (ClinicalTrials.gov NCT02289326). NIH Clinical Trials. https://clinicaltrials.gov/study/NCT02289326

  81. Ricker CA et al. (2013). A Pilot Study of Metformin in Patients With a Diagnosis of Li-Fraumeni Syndrome (ClinicalTrials.gov NCT01981525). NIH Clinical Trials. https://clinicaltrials.gov/study/NCT01981525

  82. Wang J et al. (2025). Mapping the current research landscape of metformin in cancer chemoprevention and therapy. Pharmacological Research. https://pmc.ncbi.nlm.nih.gov/articles/PMC12339854/

  83. Kannan S et al. (2020). Therapeutic Editing of the TP53 Gene: Is CRISPR/Cas9 an Option? International Journal of Molecular Sciences. https://pmc.ncbi.nlm.nih.gov/articles/PMC7349023/

  84. Villani A et al. (2020). Guidelines for the Li-Fraumeni and heritable TP53-related cancer syndromes. ERN GENTURIS. https://www.genturis.eu/l=deu/Assets/TP53-Surveillance-Guideline---ERN-GENTURIS.pdf

  85. Oronsky B et al. (2024). The impact of metformin on clinical outcomes of metastatic colorectal cancer. Journal of Clinical Oncology. https://ascopubs.org/doi/10.1200/JCO.2024.42.16_suppl.e15580

  86. Qu J et al. (2023). Arsenic trioxide extends survival of Li–Fraumeni syndrome mouse models by rescuing mutant p53. Cell Death & Disease. https://www.nature.com/articles/s41419-023-06281-2

  87. Wang X et al. (2024). TP53 Mutation-Mediated Immune Evasion in Cancer: Mechanisms and Therapeutic Implications. Cancers. https://pmc.ncbi.nlm.nih.gov/articles/PMC11393945/

  88. Mai PL et al. (2024). Forming International Collaborative Efforts to Assess Metformin Use in Li-Fraumeni Syndrome. Cancer Network. https://www.cancernetwork.com/view/forming-international-collaborative-efforts-to-assess-metformin-use-in-lfs

  89. Oren M et al. (2017). Li-Fraumeni syndrome disease model: A platform to develop precision cancer therapy. Trends in Molecular Medicine. https://pmc.ncbi.nlm.nih.gov/articles/PMC5752137/

  90. Wang H et al. (2025). Cancer chemoprevention: signaling pathways and strategic interventions. Signal Transduction and Targeted Therapy. https://www.nature.com/articles/s41392-025-02167-1

  91. Levine AJ et al. (2025). Therapeutic potential of combating cancer by restoring wild-type p53 function through mRNA nanomedicine. Current Opinion in Oncology. https://www.sciencedirect.com/science/article/pii/S1549963424000017

  92. FORCE (2025). Cancer Risk Management for People with an Inherited TP53 Mutation. Facing Our Risk of Cancer Empowered. https://www.facingourrisk.org/info/hereditary-cancer-and-genetic-testing/hereditary-cancer-genes-and-risk/genes-by-name/tp53/ris

  93. The George Pantziarka TP53 Trust (2024). Active Trials and Projects - MILI Trial. TP53 Trust. https://www.tp53.co.uk/projects/

  94. Swisher SG et al. (2003). p53 therapy in a patient with Li-Fraumeni syndrome. Molecular Cancer Therapeutics. https://aacrjournals.org/mct/article/6/5/1478/235084/p53-therapy-in-a-patient-with-Li-Fraumeni-syndrome

  95. Pinto EM et al. (2020). XAF1 as a modifier of p53 function and cancer susceptibility. Science Advances (Mayo Clinic). https://mayoclinic.elsevierpure.com/en/publications/xaf1-as-a-modifier-of-p53-function-and-cancer-susceptibility/

  96. Huang C et al. (2017). Functional consequence of the p53 codon 72 polymorphism in colorectal cancer. Oncotarget. https://pmc.ncbi.nlm.nih.gov/articles/PMC5652727/

  97. Pinto EM et al. (2020). XAF1 as a modifier of p53 function and cancer susceptibility. Science Advances. https://pmc.ncbi.nlm.nih.gov/articles/PMC7314530/

  98. Murphy ME (2019). Dynamic role of the codon 72 p53 single-nucleotide polymorphism in tumor formation. Oncogene. https://www.nature.com/articles/s41388-018-0630-4

  99. Spector LG et al. (2016). Genome-Wide DNA Methylation Analysis Reveals Epigenetic Dysregulation in Peripheral Blood Cells from Li-Fraumeni Syndrome Patients. Journal of Clinical Oncology. https://pmc.ncbi.nlm.nih.gov/articles/PMC6366343/

  100. Pinto EM et al. (2020). XAF1 as a modifier of p53 function and cancer susceptibility. UCL Discovery Repository. https://discovery.ucl.ac.uk/id/eprint/10105076/

  101. Azzam GA et al. (2003). TP53 codon 72 Gene Polymorphism Paradox in Associated Cancer Risk. Asian Pacific Journal of Cancer Prevention. https://pmc.ncbi.nlm.nih.gov/articles/PMC3614723/

  102. Pinto EM et al. (2020). XAF1 as a modifier of p53 function and cancer susceptibility. PubMed. https://pubmed.ncbi.nlm.nih.gov/32637605/

  103. Ruijs MW et al. (2011). Sex-specific effect of the TP53 PIN3 polymorphism on cancer risk in TP53 mutation carriers. Journal of Medical Genetics. https://pmc.ncbi.nlm.nih.gov/articles/PMC6497528/

  104. Slavin TP et al. (2025). Cancer risk in carriers of TP53 germline variants grouped into functionally defined classes. JNCI Cancer Spectrum. https://academic.oup.com/jncics/article/9/1/pkaf008/7985583

  105. Pinto EM et al. (2023). Multiple TP53 p.R337H haplotypes and implications for tumor susceptibility. Cell Genomics. https://www.sciencedirect.com/science/article/pii/S2666247723000763

  106. Murphy ME (2019). Common genetic variants in the TP53 pathway and their impact on cancer. Journal of Molecular Cell Biology. https://academic.oup.com/jmcb/article/11/7/578/5509869

  107. Custodio G et al. (2020). What 20 years of research has taught us about the TP53 p.R337H mutation. Cancer. https://acsjournals.onlinelibrary.wiley.com/doi/full/10.1002/cncr.33143

  108. Heidari M et al. (2021). Genetic associations of TP53 codon Pro72Arg polymorphism with coronary artery disease. Gene Reports. https://www.sciencedirect.com/science/article/abs/pii/S2452014421001618

  109. Berisha S et al. (2025). Peripheral blood DNA methylation predicts the early onset of cancer in Li-Fraumeni syndrome. Clinical Cancer Research. https://pmc.ncbi.nlm.nih.gov/articles/PMC12381258/

  110. Correa H et al. (2022). Li–Fraumeni-associated pancreatic neuroendocrine tumour and XAF1 p.E134* risk modifier. ecancermedicalscience. https://ecancer.org/en/journal/article/1487-li-fraumeni-associated-pancreatic-neuroendocrine-tumour-and-xaf1-p-glu134ter-risk-mo

  111. Zhu G et al. (2021). Effect of the p53 P72R Polymorphism on Mutant TP53 Allele Selection in Human Cancer. JNCI. https://academic.oup.com/jnci/article/113/9/1246/6130823

  112. Poruk KE et al. (2025). TP53 Mutations May Reveal Viability of Different Pancreatic Cancer Therapies. Cancer Network. https://www.cancernetwork.com/view/p53-mutations-may-reveal-viability-of-different-pancreatic-cancer-therapies

  113. BioPharma Reporter (2025). Pancreatic cancer: The clinical trial landscape and progress for 2024. Clinical Trials Arena. https://www.clinicaltrialsarena.com/sponsored/pancreatic-cancer-the-clinical-trial-landscape-and-progress-for-2024/

  114. Koong AC et al. (2022). The Landscape of Genetic Alterations Based on TP53 Status in Pancreatic Adenocarcinoma After Radiation Therapy. International Journal of Radiation Oncology Biology Physics. https://www.sciencedirect.com/science/article/abs/pii/S0360301622013657

  115. Singhi AD et al. (2025). Evaluating the Effect of KRAS Variants on Survival Outcomes in Metastatic Pancreatic Ductal Adenocarcinoma. Journal of Clinical Oncology. https://pmc.ncbi.nlm.nih.gov/articles/PMC12379785/

  116. UCSD Health (2025). UCSD Pancreatic Cancer Clinical Trials for 2025. UC San Diego Clinical Trials. https://clinicaltrials.ucsd.edu/pancreatic-cancer

  117. Gemenetzis G et al. (2023). TP53 Gain-of-Function and Non–Gain-of-Function Mutations Are Differentially Associated With Survival in Metastatic Pancreatic Adenocarcinoma. JCO Precision Oncology. https://ascopubs.org/doi/10.1200/PO.22.00570

  118. Singhi AD et al. (2025). KRAS Mutation Status and Outcomes in Metastatic Pancreatic Cancer. JAMA Network Open. https://jamanetwork.com/journals/jamanetworkopen/fullarticle/2828753

  119. Novotech CRO (2025). Pancreatic Cancer: Global Clinical Trial Landscape (2024). Novotech Reports. https://novotech-cro.com/reports/pancreatic-cancer-global-clinical-trial-landscape-2024

  120. Jiang Z et al. (2021). TP53 Mutational Status-Based Genomic Signature for Prognosis in Pancreatic Cancer. Frontiers in Cell and Developmental Biology. https://www.frontiersin.org/journals/cell-and-developmental-biology/articles/10.3389/fcell.2021.665265/full

  121. Singhi AD et al. (2023). Precision medicine for KRAS wild-type pancreatic adenocarcinomas. European Journal of Cancer. https://www.sciencedirect.com/science/article/pii/S0959804923007992

  122. Ferrone CR et al. (2025). Treatment Innovations in Pancreatic Cancer: Putting Patient Outcomes First. ASCO Educational Book. https://ascopubs.org/doi/10.1200/EDBK-25-473204

  123. Singhi AD et al. (2020). Alterations in Driver Genes Predict Survival in Patients with Resected Pancreatic Ductal Adenocarcinoma. Memorial Sloan Kettering Cancer Center. https://www.mskcc.org/clinical-updates/alterations-driver-genes-predict-survival-patients-resected-pancreatic-ductal-adenocarcin

  124. Maker AV et al. (2018). A KRAS wild type mutational status confers a survival advantage in pancreatic ductal adenocarcinoma. Journal for ImmunoTherapy of Cancer. https://pmc.ncbi.nlm.nih.gov/articles/PMC5848049/

  125. Singhi AD et al. (2024). Unveiling the Promise: Navigating Clinical Trials 1978–2024 for PDAC. Journal of Clinical Medicine. https://pmc.ncbi.nlm.nih.gov/articles/PMC11544830/

  126. Muller PA et al. (2022). Wild type and gain of function mutant TP53 can regulate the sensitivity of pancreatic cancer cells to chemotherapeutic drugs. Aging. https://www.aging-us.com/article/204038/text

  127. Li F et al. (2021). Complete disease remission in a TP53 and KRAS co-mutated brain oligometastatic lung cancer patient. Translational Lung Cancer Research. https://tlcr.amegroups.org/article/view/52393/html

  128. Pancreatic Cancer Action Network (2025). Clinical Trials - Pancreatic Cancer. PanCAN. https://pancan.org/facing-pancreatic-cancer/treatment/treatment-types/clinical-trials/

  129. Fisher WE et al. (2017). A Long-Term Survivor of Metastatic Pancreatic Adenocarcinoma Free of Recurrence 12 Years After Treatment. Cureus. https://www.cureus.com/articles/5660-a-long-term-survivor-of-metastatic-pancreatic-adenocarcinoma-free-of-recurrence-12-years-af

  130. Zhang J et al. (2025). Structural dynamics of the wild-type p53 DNA-binding domain and hotspot mutants reveal oncogenic conformational shifts. Physical Chemistry Chemical Physics. https://pubs.rsc.org/en/content/articlelanding/2025/cp/d5cp01257k

  131. Freed-Pastor WA et al. (2022). Different hotspot p53 mutants exert distinct phenotypes and predict outcome in colorectal cancer. Nature Communications. https://pmc.ncbi.nlm.nih.gov/articles/PMC9120190/

  132. Bell S et al. (2017). Evidence for allosteric effects on p53 oligomerization induced by phosphorylation. Protein Science. https://pmc.ncbi.nlm.nih.gov/articles/PMC5775181/

  133. Hussain T et al. (2025). Mutational Disruption of TP53: A Structural Approach to Understanding Cancer-Associated Variants. Cancers. https://pmc.ncbi.nlm.nih.gov/articles/PMC12470219/

  134. Sharath A et al. (2022). Gene Regulation by p53 in Human Cancer System. Asian Pacific Journal of Cancer Biology. http://waocp.com/journal/index.php/apjcb/article/view/835

  135. Guarracino DA et al. (2008). Stability and structural recovery of the tetramerization domain of p53-R337H mutant induced by a designed templating ligand. PNAS. https://www.pnas.org/doi/10.1073/pnas.0805658105

  136. Eldar A et al. (2013). Structural studies of p53 inactivation by DNA-contact mutations and its rescue by suppressor mutations. Nucleic Acids Research. https://academic.oup.com/nar/article/41/18/8748/1032655

  137. Ren YA et al. (2015). TP53 hot spot mutations in ovarian cancer. Journal of Ovarian Research. https://pmc.ncbi.nlm.nih.gov/articles/PMC5303002/

  138. Levine AJ (2020). p53 tetramerization: at the center of the dominant-negative effect of mutant p53. Genes & Development. https://pmc.ncbi.nlm.nih.gov/articles/PMC7462067/

  139. Hong B et al. (2023). Targeting p53 pathways: mechanisms, structures and advances in therapy. Signal Transduction and Targeted Therapy. https://www.nature.com/articles/s41392-023-01347-1

  140. Olivier M et al. (2017). Why are there hotspot mutations in the TP53 gene in human cancers? Cell Death & Differentiation. https://www.nature.com/articles/cdd2017180

  141. Gomes J et al. (2025). In-frame germline TP53 variant impairs p53 oligomerization and DNA binding. Scientific Reports. https://www.nature.com/articles/s41598-025-14684-8

  142. Ukkola A et al. (2025). Cancer Classification Through p53 Hotspot Mutations: An Ensemble Learning Approach. Cellular Biochemistry and Function. https://onlinelibrary.wiley.com/doi/10.1002/cbin.70041

  143. Schuster-Böckler B et al. (2022). Structural Basis of Mutation-Dependent p53 Tetramerization Deficiency. bioRxiv. https://www.biorxiv.org/content/10.1101/2022.05.13.491836v2.full-text

  144. Nikolova PV et al. (2000). Mechanism of rescue of common p53 cancer mutations by second-site suppressor mutations. EMBO Journal. https://www.embopress.org/doi/10.1093/emboj/19.3.370

  145. Yu X et al. (2023). Discovery of compounds that reactivate p53 mutants in vitro and in vivo. eBioMedicine. https://www.sciencedirect.com/science/article/pii/S2451945622002756

  146. Natan E et al. (2023). How Do Cancer-Related Mutations Affect the Oligomerisation State of p53? Journal of Molecular Biology. https://pdfs.semanticscholar.org/4a4c/7bed5b8e271ca58c4fe5e45e82790a15a811.pdf

  147. Bullock AN et al. (2003). Understanding the function–structure and function–mutation relationships of p53 tumor suppressor. PNAS. https://www.pnas.org/doi/10.1073/pnas.1431692100

  148. Bewersdorf JP et al. (2023). Are TP53 mutations all alike? Hematology ASH Education Program. https://ashpublications.org/hematology/article/2024/1/321/526232/Are-TP53-mutations-all-alike

  149. Soto M et al. (2023). Whole-genome doubling drives oncogenic loss of chromatin segregation. Nature. https://www.nature.com/articles/s41586-023-05794-2

  150. Malcikova J et al. (2014). Detailed analysis of therapy-driven clonal evolution of TP53 mutations in chronic lymphocytic leukemia. Leukemia. https://pmc.ncbi.nlm.nih.gov/articles/PMC4396398/

  151. Keum N et al. (2021). High prevalence of TP53 loss and whole-genome doubling in early-onset colorectal cancer. Experimental & Molecular Medicine. https://pmc.ncbi.nlm.nih.gov/articles/PMC8080557/

  152. Cousineau I et al. (2021). TP53 loss initiates chromosomal instability in fallopian tube epithelial cells. Cell Death & Differentiation. https://pmc.ncbi.nlm.nih.gov/articles/PMC8649171/

  153. Zack TI et al. (2018). Genome doubling shapes the evolution and prognosis of advanced cancers. Nature Genetics. https://pmc.ncbi.nlm.nih.gov/articles/PMC6072608/

  154. Cousineau I et al. (2021). TP53 loss initiates chromosomal instability in high-grade serous ovarian cancer precursors. bioRxiv. https://www.biorxiv.org/content/10.1101/2021.03.12.435079.full

  155. Xing D et al. (2022). Intratumor genetic heterogeneity and clonal evolution to decode cancer progression in endometrial carcinoma. Oncogene. https://www.nature.com/articles/s41388-022-02221-0

  156. Takeda DY et al. (2025). Ongoing genome doubling shapes evolvability and immunity in cancer. Nature. https://www.nature.com/articles/s41586-025-09240-3

  157. Wang X et al. (2024). Mixed responses to targeted therapy driven by chromosomal instability in TP53-mutant lung cancer. Nature Communications. https://www.nature.com/articles/s41467-024-47606-9

  158. Janku F et al. (2025). Clonal evolution and therapy resistance in the era of precision oncology. Seminars in Cancer Biology. https://www.sciencedirect.com/science/article/pii/S1044579X25001257

  159. Quinton RJ et al. (2023). Whole-Genome Doubling as a source of cancer: how, when, where, and why? Frontiers in Cell and Developmental Biology. https://www.frontiersin.org/journals/cell-and-developmental-biology/articles/10.3389/fcell.2023.1209136/full

  160. Smith JC et al. (2020). Whole genome analysis identifies the association of TP53 with 17p loss and chromosomal instability in colorectal cancer. Cancers. https://pmc.ncbi.nlm.nih.gov/articles/PMC7081316/

  161. Kuipers J et al. (2021). Single-cell and single-variant resolution analysis of clonal evolution in myeloproliferative neoplasms. bioRxiv. https://www.biorxiv.org/content/10.1101/2020.12.30.424907.full

  162. Kim J et al. (2025). The heterogeneous role of whole-genome doubling in various cancers. BMB Reports. https://www.bmbreports.org/view.html?uid=1972&vmd=Full

  163. Donehower LA et al. (2019). Integrated Analysis of TP53 Gene and Pathway Alterations in The Cancer Genome Atlas. Cell Reports. https://www.sciencedirect.com/science/article/pii/S221112471930885X

  164. Ma G et al. (2017). Mutation landscape and intra-tumor heterogeneity of two MANECs of the esophagus revealed by multi-region sequencing. Oncotarget. https://www.oncotarget.com/article/18678/text/

  165. Storchova Z et al. (2024). Cancer Genomes Sometimes Take the Longest Evolutionary Road. Cancer Discovery. https://aacrjournals.org/cancerdiscovery/article/14/10/1766/748595/Cancer-Genomes-Sometimes-Take-the-Longest

  166. Levine AJ et al. (2010). Links Between Mutant p53 and Genomic Instability. Journal of Cellular Biochemistry. https://pmc.ncbi.nlm.nih.gov/articles/PMC4407809/

Sources [1] Analysis of the Li-Fraumeni Spectrum Based on an International ... https://jamanetwork.com/journals/jamaoncology/fullarticle/2785678 [2] Multiple TP53 p.R337H haplotypes and implications for tumor ... https://pmc.ncbi.nlm.nih.gov/articles/PMC10597792/ [3] Cancer associated to low-penetrance TP53 variants - PubMed https://pubmed.ncbi.nlm.nih.gov/40268607/ [4] Why are there hotspot mutations in the TP53 gene in human cancers? https://pmc.ncbi.nlm.nih.gov/articles/PMC5729536/ [5] Structural dynamics of the wild-type p53 DNA-binding domain and ... https://pubs.rsc.org/en/content/articlelanding/2025/cp/d5cp01257k [6] Why are there hotspot mutations in the TP53 gene in human cancers? https://www.nature.com/articles/cdd2017180 [7] Targeted point mutations of p53 lead to dominant-negative ... - PNAS https://www.pnas.org/doi/10.1073/pnas.052713099 [8] Structural studies of p53 inactivation by DNA-contact mutations and ... https://academic.oup.com/nar/article/41/18/8748/1032655 [9] Different hotspot p53 mutants exert distinct phenotypes and predict ... https://pmc.ncbi.nlm.nih.gov/articles/PMC9120190/ [10] The Common Germline TP53-R337H Mutation Is Hypomorphic and ... https://aacrjournals.org/cancerres/article/81/9/2442/670613/The-Common-Germline-TP53-R337H-Mutation-Is [11] What 20 years of research has taught us about the TP53 p.R337H ... https://acsjournals.onlinelibrary.wiley.com/doi/full/10.1002/cncr.33143 [12] Stability and structural recovery of the tetramerization domain of p53 ... https://www.pnas.org/doi/10.1073/pnas.0805658105 [13] at the center of the dominant-negative effect of mutant p53 https://genesdev.cshlp.org/content/34/17-18/1128.full [14] A dominant-negative effect drives selection of TP53 missense ... https://www.science.org/doi/10.1126/science.aax3649 [15] A dominant-negative effect drives selection of TP53 missense ... - NIH https://pmc.ncbi.nlm.nih.gov/articles/PMC7327437/ [16] Revisiting tumor patterns and penetrance in germline TP53 mutation ... https://pubmed.ncbi.nlm.nih.gov/29076966/ [17] Are There Low-Penetrance TP53 Alleles? Evidence from Childhood ... https://pmc.ncbi.nlm.nih.gov/articles/PMC1288270/ [18] XAF1 as a modifier of p53 function and cancer susceptibility https://mayoclinic.elsevierpure.com/en/publications/xaf1-as-a-modifier-of-p53-function-and-cancer-susceptibility/ [19] XAF1 as a modifier of p53 function and cancer susceptibility - NIH https://pmc.ncbi.nlm.nih.gov/articles/PMC7314530/ [20] Functional consequence of the p53 codon 72 polymorphism in ... - NIH https://pmc.ncbi.nlm.nih.gov/articles/PMC5652727/ [21] Dynamic role of the codon 72 p53 single-nucleotide polymorphism ... https://www.nature.com/articles/s41388-018-0630-4 [22] TP53 codon 72 Gene Polymorphism Paradox in Associated ... - NIH https://pmc.ncbi.nlm.nih.gov/articles/PMC3614723/ [23] Sex-specific effect of the TP53 PIN3 polymorphism on cancer risk in ... https://pmc.ncbi.nlm.nih.gov/articles/PMC6497528/ [24] Cell-free DNA from germline TP53 mutation carriers reflect cancer ... https://www.nature.com/articles/s41467-024-51529-w [25] Genome-Wide DNA Methylation Analysis Reveals Epigenetic ... https://pmc.ncbi.nlm.nih.gov/articles/PMC6366343/ [26] TP63, TP73: The Guardian's Elder Brothers - NCBI - NIH https://www.ncbi.nlm.nih.gov/books/NBK6163/ [27] Functional interplay between MDM2, p63/p73 and mutant p53 - Nature https://www.nature.com/articles/onc2014359 [28] MDM2, MDMX, and p73 regulate cell-cycle progression in ... - PNAS https://www.pnas.org/doi/10.1073/pnas.2102420118 [29] Tumor Suppressor p53: Biology, Signaling Pathways, and ... https://pmc.ncbi.nlm.nih.gov/articles/PMC8730328/ [30] Recent Research on Role of p53 Family in Small-Cell Lung Cancer https://pmc.ncbi.nlm.nih.gov/articles/PMC11988120/ [31] Clinicogenomic landscape of pancreatic adenocarcinoma identifies ... https://www.nature.com/articles/s41591-024-03362-3 [32] Clinical and genetic characteristics of carriers of the TP53 c.541C ... https://pmc.ncbi.nlm.nih.gov/articles/PMC11512851/ [33] 1349-TP53 (Li-Fraumeni) – risk management (child) - eviQ https://www.eviq.org.au/cancer-genetics/paediatric/risk-management/1349-tp53-li-fraumeni-risk-management-child [34] Gene Regulation by p53 in Human Cancer System http://waocp.com/journal/index.php/apjcb/article/view/835 [35] Cancer Precision-Prevention trial of Metformin in adults with Li ... - NIH https://pmc.ncbi.nlm.nih.gov/articles/PMC10837926/ [36] Clonal dynamics following p53 loss of heterozygosity in Kras-driven ... https://www.nature.com/articles/ncomms12685 [37] Ordered and deterministic cancer genome evolution after p53 loss https://www.nature.com/articles/s41586-022-05082-5 [38] Loss of p53 tumor suppression function drives invasion ... - bioRxiv https://www.biorxiv.org/content/10.1101/2021.12.31.472823v1.full-text [39] Mutant p53 drives metastasis and overcomes growth arrest ... - PNAS https://www.pnas.org/doi/10.1073/pnas.0908428107 [40] Ordered and deterministic cancer genome evolution after p53 loss https://pmc.ncbi.nlm.nih.gov/articles/PMC9402436/ [41] Genome doubling shapes the evolution and prognosis of advanced ... https://pmc.ncbi.nlm.nih.gov/articles/PMC6072608/ [42] Pan-cancer inference of intra-tumor heterogeneity reveals ... https://journals.plos.org/plosgenetics/article?id=10.1371%2Fjournal.pgen.1007669 [43] TP53 loss initiates chromosomal instability in fallopian tube ... - NIH https://pmc.ncbi.nlm.nih.gov/articles/PMC8649171/ [44] TP53 loss initiates chromosomal instability in high-grade serous ... https://www.biorxiv.org/content/10.1101/2021.03.12.435079.full [45] Gradual Telomere Shortening and Increasing Chromosomal ... https://journals.plos.org/plosone/article?id=10.1371%2Fjournal.pone.0117575 [46] Chromosomal Instability as a Driver of Tumor Heterogeneity and ... https://pmc.ncbi.nlm.nih.gov/articles/PMC5453382/ [47] Mixed responses to targeted therapy driven by chromosomal ... https://www.nature.com/articles/s41467-024-47606-9 [48] Whole-genome doubling drives oncogenic loss of chromatin ... https://www.nature.com/articles/s41586-023-05794-2 [49] Ongoing genome doubling shapes evolvability and immunity in ... https://www.nature.com/articles/s41586-025-09240-3 [50] Whole-Genome Doubling as a source of cancer - Frontiers https://www.frontiersin.org/journals/cell-and-developmental-biology/articles/10.3389/fcell.2023.1209136/full [51] SMAD4 Gene Mutations Are Associated With Poor Prognosis ... - PMC https://pmc.ncbi.nlm.nih.gov/articles/PMC2819274/ [52] Very long-term survival in pancreatic cancer - PMC - NIH https://pmc.ncbi.nlm.nih.gov/articles/PMC4505162/ [53] Significance of TP53, CDKN2A, SMAD4 and KRAS in Pancreatic ... https://pmc.ncbi.nlm.nih.gov/articles/PMC11049225/ [54] TP53 Gain-of-Function and Non–Gain-of-Function Mutations Are ... https://ascopubs.org/doi/10.1200/PO.22.00570 [55] TP53 Mutations May Reveal Viability of Different Pancreatic Cancer ... https://www.cancernetwork.com/view/p53-mutations-may-reveal-viability-of-different-pancreatic-cancer-therapies [56] Evaluating the Effect of KRAS Variants on Survival Outcomes ... - NIH https://pmc.ncbi.nlm.nih.gov/articles/PMC12379785/ [57] Precision medicine for KRAS wild-type pancreatic adenocarcinomas https://www.sciencedirect.com/science/article/pii/S0959804923007992 [58] KRAS Mutation Status and Outcomes in Metastatic Pancreatic ... https://jamanetwork.com/journals/jamanetworkopen/fullarticle/2828753 [59] Immune infiltrates as predictive markers of survival in pancreatic ... https://pmc.ncbi.nlm.nih.gov/articles/PMC3738865/ [60] Distinct immune cell infiltration patterns in pancreatic ductal ... - Nature https://www.nature.com/articles/s41467-024-55424-2 [61] Immune cell infiltrates as prognostic biomarkers in pancreatic ductal ... https://pmc.ncbi.nlm.nih.gov/articles/PMC7869931/ [62] Exceptional responders to immunotherapy in pancreatic cancer - NIH https://pmc.ncbi.nlm.nih.gov/articles/PMC12151408/ [63] Risk and prognostic factors of survival for patients with pancreatic ... https://www.frontiersin.org/journals/oncology/articles/10.3389/fonc.2025.1521616/full [64] It Is All in the Genes: A Story of Unexpected Survival in a 67-Year ... https://pmc.ncbi.nlm.nih.gov/articles/PMC10400294/ [65] NCT01981525 | A Pilot Study of Metformin in Patients With a ... https://clinicaltrials.gov/study/NCT01981525 [66] Mapping the current research landscape of metformin in cancer ... https://pmc.ncbi.nlm.nih.gov/articles/PMC12339854/ [67] Cancer chemoprevention: signaling pathways and strategic ... - Nature https://www.nature.com/articles/s41392-025-02167-1 [68] Forming International Collaborative Efforts to Assess Metformin Use ... https://www.cancernetwork.com/view/forming-international-collaborative-efforts-to-assess-metformin-use-in-lfs [69] Active Trials and Projects - The George Pantziarka TP53 Trust https://www.tp53.co.uk/projects/ [70] Therapeutic Editing of the TP53 Gene: Is CRISPR/Cas9 an Option? https://pmc.ncbi.nlm.nih.gov/articles/PMC7349023/ [71] Therapeutic potential of combating cancer by restoring wild-type p53 ... https://www.sciencedirect.com/science/article/pii/S1549963424000017 [72] Li-Fraumeni syndrome disease model: A platform to develop ... https://pmc.ncbi.nlm.nih.gov/articles/PMC5752137/ [73] p53 Prophylactic Therapy for Cancer Prevention - LFS Association https://www.lfsassociation.org/lfs-news/p53-prophylactic-therapy-for-cancer-prevention/ [74] Arsenic trioxide extends survival of Li–Fraumeni syndrome ... - Nature https://www.nature.com/articles/s41419-023-06281-2 [75] p53 therapy in a patient with Li-Fraumeni syndrome - AACR Journals https://aacrjournals.org/mct/article/6/5/1478/235084/p53-therapy-in-a-patient-with-Li-Fraumeni-syndrome [76] TP53 Mutation-Mediated Immune Evasion in Cancer - PubMed Central https://pmc.ncbi.nlm.nih.gov/articles/PMC11393945/ [77] Treatment Innovations in Pancreatic Cancer: Putting Patient ... https://ascopubs.org/doi/10.1200/EDBK-25-473204 [78] Study Details | Biomarker Monitoring in TP53 Mutation Carriers https://clinicaltrials.gov/study/NCT02289326 [79] [PDF] guidelines for the li-fraumeni - ERN GENTURIS https://www.genturis.eu/l=deu/Assets/TP53-Surveillance-Guideline---ERN-GENTURIS.pdf [80] Guidelines for the Li–Fraumeni and heritable TP53-related cancer ... https://www.nature.com/articles/s41431-020-0638-4 [81] The Role of TP53 Mutations in the Origin and Evolution of Therapy ... https://pmc.ncbi.nlm.nih.gov/articles/PMC4403236/ [82] Heterogeneity of TP53 Mutations and P53 Protein Residual Function ... https://pmc.ncbi.nlm.nih.gov/articles/PMC7655923/ [83] Emerging insights into ethnic-specific TP53 germline variants | JNCI https://academic.oup.com/jnci/article/115/10/1145/7205922 [84] TP53_PROF: a machine learning model to predict impact of ... https://academic.oup.com/bib/article/23/2/bbab524/6510957 [85] Cancer associated to low-penetrance TP53 variants: insights from p ... https://www.sciencedirect.com/science/article/abs/pii/S2405803325000950 [86] Clinicopathologic Features of Breast Tumors in Germline TP53 ... https://www.ovid.com/journals/ajsp/pdf/10.1097/pas.0000000000002338~clinicopathologic-features-of-breast-tumors-in-germline-tp53 [87] TP53 variant clusters stratify phenotypic diversity in germline carriers ... https://www.nature.com/articles/s41467-025-63528-6 [88] [PDF] Variation at the R181 residue of p53 confers loss of p53 DNA ... https://www.biorxiv.org/content/10.1101/2025.08.12.669925v1.full.pdf [89] TP53: the unluckiest of genes? | Cell Death & Differentiation - Nature https://www.nature.com/articles/s41418-024-01391-6 [90] Mutational Landscape of TP53 Across Cancer Types https://sciety-discovery.elifesciences.org/articles/by?article_doi=10.1101%2F2025.08.12.669884 [91] Deciphering the significance of p53 mutant proteins - ScienceDirect https://www.sciencedirect.com/science/article/pii/S096289242400117X [92] Targeting p53 for the treatment of cancer - ScienceDirect.com https://www.sciencedirect.com/science/article/pii/S1044579X20301607 [93] TP53 variant clusters stratify phenotypic diversity in germline carriers ... https://pmc.ncbi.nlm.nih.gov/articles/PMC12480935/ [94] TP53 Testing and Li-Fraumeni Syndrome: Current Status of Clinical ... https://pmc.ncbi.nlm.nih.gov/articles/PMC3627545/ [95] The TP53 tumor suppressor gene: From molecular biology to clinical ... https://onlinelibrary.wiley.com/doi/10.1111/joim.20106?af=R [96] Personalized screening strategies for TP53 R337H carriers: a ... https://www.sciencedirect.com/science/article/pii/S2667193X24003090 [97] Allele-specific wild-type TP53 expression in the unaffected carrier ... https://www.sciencedirect.com/science/article/abs/pii/S2210776217300066 [98] AI-guided identification of risk variants for adrenocortical tumours in ... https://www.thelancet.com/journals/lanam/article/PIIS2667-193X(24)00190-X/fulltext [99] carcinogenic mechanisms in breast cancer with TP53 pathogenic ... https://pmc.ncbi.nlm.nih.gov/articles/PMC11341599/ [100] Are There Low-Penetrance TP53 Alleles? Evidence from Childhood ... https://www.sciencedirect.com/science/article/pii/S0002929707626020 [101] Loss of p53 function at late stages of tumorigenesis confers ARF ... https://www.pnas.org/doi/10.1073/pnas.1910255116 [102] The Role of p53 Mutations in Early and Late Response to Mitotic ... https://pmc.ncbi.nlm.nih.gov/articles/PMC11852650/ [103] Genomic Instability in Cancer: Teetering on the Limit of Tolerance https://aacrjournals.org/cancerres/article/77/9/2179/625089/Genomic-Instability-in-Cancer-Teetering-on-the [104] Trp53R172H and KrasG12D cooperate to promote chromosomal ... https://www.sciencedirect.com/science/article/pii/S1535610805001285 [105] Concurrent TP53 Mutations Facilitate Resistance Evolution in EGFR ... https://www.sciencedirect.com/science/article/pii/S1556086422001411 [106] Chromosome instability and carcinogenesis: Insights from murine ... https://febs.onlinelibrary.wiley.com/doi/10.1016/j.molonc.2013.10.005 [107] Clonal Heterogeneity and Tumor Evolution: Past, Present, and the ... https://www.sciencedirect.com/science/article/pii/S0092867417300661 [108] Mutant p53 in cancer therapy—the barrier or the path https://academic.oup.com/jmcb/article/11/4/293/5227720 [109] Mutations of p53 associated with pancreatic cancer and therapeutic ... https://pmc.ncbi.nlm.nih.gov/articles/PMC8382872/ [110] Abstract 6217: TP53 loss with whole genome doubling mediates ... https://aacrjournals.org/cancerres/article/82/12_Supplement/6217/703805/Abstract-6217-TP53-loss-with-whole-genome-doubling [111] Clinico-pathological analyses of the relationship between SMAD4 ... https://ascopubs.org/doi/10.1200/JCO.2018.36.4_suppl.319 [112] Clinical Effect of Driver Mutations of KRAS, CDKN2A/P16, TP53, and ... https://www.liebertpub.com/doi/10.1089/gtmb.2020.0078 [113] A quantitative score of immune cell infiltration predicts the prognosis ... https://www.sciencedirect.com/science/article/abs/pii/S1567576921005269 [114] prognostic significance of TP53 status in gemcitabine plus nab ... https://www.sciencedirect.com/science/article/pii/S2949819825000901 [115] New insight into the role of SMAD4 mutation/deficiency in the ... https://www.sciencedirect.com/science/article/abs/pii/S0304419X24001513 [116] Molecular characterization and prognostic implications of KRAS ... https://www.nature.com/articles/s41698-025-01087-1 [117] Prediction of overall survival in pancreatic cancer based on a ... - NIH https://pmc.ncbi.nlm.nih.gov/articles/PMC12091656/ [118] Long-term survivors of pancreatic adenocarcinoma show low rates ... https://journals.sagepub.com/doi/10.3233/CBM-170464 [119] Post‐operative mortality and recurrence patterns in pancreatic ... https://pathsocjournals.onlinelibrary.wiley.com/doi/full/10.1002/cjp2.323 [120] The impact of metformin on clinical outcomes of metastatic colorectal ... https://ascopubs.org/doi/10.1200/JCO.2024.42.16_suppl.e15580 [121] Cancer Risk Management for People with an Inherited TP53 Mutation https://www.facingourrisk.org/info/hereditary-cancer-and-genetic-testing/hereditary-cancer-genes-and-risk/genes-by-name/tp53/risk-management [122] XAF1 as a modifier of p53 function and cancer susceptibility https://discovery.ucl.ac.uk/id/eprint/10105076/ [123] XAF1 as a modifier of p53 function and cancer susceptibility - PubMed https://pubmed.ncbi.nlm.nih.gov/32637605/ [124] Cancer risk in carriers of TP53 germline variants grouped into ... https://academic.oup.com/jncics/article/9/1/pkaf008/7985583 [125] Article Multiple TP53 p.R337H haplotypes and implications for tumor ... https://www.sciencedirect.com/science/article/pii/S2666247723000763 [126] Common genetic variants in the TP53 pathway and their impact on ... https://academic.oup.com/jmcb/article/11/7/578/5509869 [127] Genetic associations of TP53 codon Pro72Arg polymorphism ... https://www.sciencedirect.com/science/article/abs/pii/S2452014421001618 [128] Peripheral blood DNA methylation predicts the early onset of ... - NIH https://pmc.ncbi.nlm.nih.gov/articles/PMC12381258/ [129] Li–Fraumeni-associated pancreatic neuroendocrine tumour and ... https://ecancer.org/en/journal/article/1487-li-fraumeni-associated-pancreatic-neuroendocrine-tumour-and-xaf1-p-glu134ter-risk-modifier-variant [130] Effect of the p53 P72R Polymorphism on Mutant TP53 Allele ... https://academic.oup.com/jnci/article/113/9/1246/6130823 [131] Pancreatic cancer: The clinical trial landscape and progress for 2024 https://www.clinicaltrialsarena.com/sponsored/pancreatic-cancer-the-clinical-trial-landscape-and-progress-for-2024/ [132] The Landscape of Genetic Alterations Based on TP53 Status in ... https://www.sciencedirect.com/science/article/abs/pii/S0360301622013657 [133] UCSD Pancreatic Cancer Clinical Trials for 2025 — San Diego https://clinicaltrials.ucsd.edu/pancreatic-cancer [134] Pancreatic Cancer: Global Clinical Trial Landscape (2024) - Novotech https://novotech-cro.com/reports/pancreatic-cancer-global-clinical-trial-landscape-2024 [135] TP53 Mutational Status-Based Genomic Signature for Prognosis ... https://www.frontiersin.org/journals/cell-and-developmental-biology/articles/10.3389/fcell.2021.665265/full [136] Alterations in Driver Genes Predict Survival in Patients with ... https://www.mskcc.org/clinical-updates/alterations-driver-genes-predict-survival-patients-resected-pancreatic-ductal-adenocarcinoma [137] A KRAS wild type mutational status confers a survival advantage in ... https://pmc.ncbi.nlm.nih.gov/articles/PMC5848049/ [138] Unveiling the Promise: Navigating Clinical Trials 1978–2024 for PDAC https://pmc.ncbi.nlm.nih.gov/articles/PMC11544830/ [139] Wild type and gain of function mutant TP53 can regulate ... - Aging-US https://www.aging-us.com/article/204038/text [140] Complete disease remission in a TP53 and KRAS co-mutated brain ... https://tlcr.amegroups.org/article/view/52393/html [141] Clinical Trials - Pancreatic Cancer Action Network https://pancan.org/facing-pancreatic-cancer/treatment/treatment-types/clinical-trials/ [142] A Long-Term Survivor of Metastatic Pancreatic Adenocarcinoma https://www.cureus.com/articles/5660-a-long-term-survivor-of-metastatic-pancreatic-adenocarcinoma-free-of-recurrence-12-years-after-treatment-of-oligometastatic-disease [143] Evidence for allosteric effects on p53 oligomerization induced by ... https://pmc.ncbi.nlm.nih.gov/articles/PMC5775181/ [144] Mutational Disruption of TP53: A Structural Approach to ... https://pmc.ncbi.nlm.nih.gov/articles/PMC12470219/ [145] TP53 hot spot mutations in ovarian cancer - PubMed Central - NIH https://pmc.ncbi.nlm.nih.gov/articles/PMC5303002/ [146] p53 tetramerization: at the center of the dominant-negative effect of ... https://pmc.ncbi.nlm.nih.gov/articles/PMC7462067/ [147] Targeting p53 pathways: mechanisms, structures and advances in ... https://www.nature.com/articles/s41392-023-01347-1 [148] In-frame germline TP53 variant impairs p53 oligomerization and ... https://www.nature.com/articles/s41598-025-14684-8 [149] Cancer Classification Through p53 Hotspot Mutations: An Ensemble ... https://onlinelibrary.wiley.com/doi/10.1002/cbin.70041 [150] Structural Basis of Mutation-Dependent p53 Tetramerization ... https://www.biorxiv.org/content/10.1101/2022.05.13.491836v2.full-text [151] Mechanism of rescue of common p53 cancer mutations by second ... https://www.embopress.org/doi/10.1093/emboj/19.3.370 [152] Discovery of compounds that reactivate p53 mutants in vitro and in ... https://www.sciencedirect.com/science/article/pii/S2451945622002756 [153] [PDF] How Do Cancer-Related Mutations Affect the Oligomerisation State ... https://pdfs.semanticscholar.org/4a4c/7bed5b8e271ca58c4fe5e45e82790a15a811.pdf [154] Understanding the function–structure and function–mutation ... - PNAS https://www.pnas.org/doi/10.1073/pnas.1431692100 [155] Are TP53 mutations all alike? | Hematology, ASH Education Program https://ashpublications.org/hematology/article/2024/1/321/526232/Are-TP53-mutations-all-alike [156] Detailed analysis of therapy-driven clonal evolution of TP53 ... https://pmc.ncbi.nlm.nih.gov/articles/PMC4396398/ [157] High prevalence of TP53 loss and whole-genome doubling in early ... https://pmc.ncbi.nlm.nih.gov/articles/PMC8080557/ [158] Intratumor genetic heterogeneity and clonal evolution to decode ... https://www.nature.com/articles/s41388-022-02221-0 [159] Clonal evolution and therapy resistance in the era of precision ... https://www.sciencedirect.com/science/article/pii/S1044579X25001257 [160] Whole genome analysis identifies the association of TP53 ... - NIH https://pmc.ncbi.nlm.nih.gov/articles/PMC7081316/ [161] Single-cell and single-variant resolution analysis of clonal evolution ... https://www.biorxiv.org/content/10.1101/2020.12.30.424907.full [162] the heterogeneous role of whole-genome doubling in various ... https://www.bmbreports.org/view.html?uid=1972&vmd=Full [163] Integrated Analysis of TP53 Gene and Pathway Alterations in The ... https://www.sciencedirect.com/science/article/pii/S221112471930885X [164] Mutation landscape and intra-tumor heterogeneity of two MANECs ... https://www.oncotarget.com/article/18678/text/ [165] Cancer Genomes Sometimes Take the Longest Evolutionary Road https://aacrjournals.org/cancerdiscovery/article/14/10/1766/748595/Cancer-Genomes-Sometimes-Take-the-Longest [166] Links Between Mutant p53 and Genomic Instability - PubMed Central https://pmc.ncbi.nlm.nih.gov/articles/PMC4407809/