Open Access

RAD51 and PALB2 in precision oncology: Clinical implications for HRD associated breast and ovarian cancers (Review)

  • Authors:
    • Mohd Adnan Kausar
    • Khalid Farhan Alshammari
    • Fahaad Alenazi
    • Sadaf Anwar
    • Amany Mohammed Khalifa
    • Tarig Ginawi
    • Abdulaziz Asiri
    • Mohammad Zeeshan Najm
    • Syed Arman Rabbani
    • Mohamed El‑Tanani
    • Saumyatika Gantayat
  • View Affiliations

  • Published online on: July 3, 2025     https://doi.org/10.3892/ijo.2025.5771
  • Article Number: 65
  • Copyright: © Kausar et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Maintaining genomic stability is essential for reducing the risk of carcinogenesis. Homologous recombination (HR) is a high‑fidelity DNA repair mechanism that addresses double‑strand breaks and interstrand crosslinks. The present review examined two key components of HR: RAD51, the eukaryotic recombinase and PALB2, a scaffolding protein. Their structural and functional roles are explored in the context of breast and ovarian cancer. RAD51 facilitates homology search and strand invasion, while PALB2 links BRCA1 and BRCA2, stabilizing RAD51 filaments. Mutations in these genes compromise HR, increasing susceptibility to various cancers and impacting treatment efficacy by impairing DNA repair. The present review discussed the clinical implications of RAD51 and PALB2 mutations, focusing on risk stratification, PARP inhibitor efficacy and emerging therapies. Additionally, it highlighted the potential of RAD51 and PALB2 as biomarkers and therapeutic targets, contributing to advances in personalized cancer management.

Introduction

Cancer develops due to genetic and epigenetic alterations that disrupt normal cellular functions, leading to uncontrolled proliferation and genomic instability. As the second leading cause of global mortality, its incidence continues to rise, driven by errors in cell division, environmental exposures (such as tobacco smoke and UV radiation) and inherited mutations (1). Among the most prevalent malignancies, breast cancer is the most frequently diagnosed cancer in women worldwide, whereas ovarian cancer, although less common, has a higher fatality rate (2). According to the World Health Organization, the global incidence of these cancers is projected to rise from 2.62 million in 2022 to 4.06 million by 2050, with mortality increasing from 873,000 to 1.49 million during the same period (3).

Genetic predisposition plays a central role in the development of these cancers, interacting with environmental and lifestyle factors. In Asia and India, mortality rates are expected to increase by 67.6 and 100.5%, respectively, by 2050, with similar trends in incidence. Although lifestyle factors contribute to cancer risk, genetic mutations, particularly in BRCA1 and BRCA2, are well-established drivers of hereditary breast and ovarian cancer syndrome (3-5). The increased mortality rates linked to breast and ovarian malignancies can mostly be ascribed to postponed diagnosis and the emergence of therapeutic resistance (6,7). Over 55% of ovarian cancer patients are identified at advanced stages (III/IV), where metastases and chemoresistance markedly restrict curative alternatives (8). Likewise, aggressive variants of breast cancer, including triple-negative breast cancer, often exhibit resistance to standard treatments such as platinum-based chemotherapy and radiotherapy, resulting in unfavorable prognoses (9,10). The evolution of resistance mechanisms entails intricate biochemical pathways that influence drug inflow and efflux, drug inactivation, DNA damage repair and apoptotic signaling. Combating delayed diagnosis via awareness campaigns and therapy resistance with targeted molecular strategies is essential for enhancing outcomes in gynecologic malignancies (6,11). Advances in next-generation sequencing have identified additional susceptibility genes, including TP53, PTEN, PALB2 and RAD51, further expanding the understanding of genetic influences on cancer risk (12-14).

RAD51 and PALB2 are key components of homologous recombination repair (HRR), a critical pathway for maintaining genomic stability. RAD51, located on chromosome 15q15.1, encodes a 339-amino acid protein essential for DNA repair, whereas PALB2, on chromosome 16p12.2, functions as a molecular scaffold, linking BRCA1 and BRCA2 to stabilize RAD51 filaments (15-20). Mutations in these genes compromise HRR, increasing susceptibility to breast, ovarian and other cancers. The present review examines their roles in cancer biology, their clinical significance and their potential as therapeutic targets (Fig. 1).

Domain organization

RAD51

As a 37-kDa protein composed of 339 amino acids in humans, RAD51 has a bipartite structure that facilitates its recombinase function. Its structure is highly conserved across species, underscoring its essential role in cellular processes. Among its functional domains, RAD51 possesses an ATPase domain responsible for DNA strand exchange and repair (21). The N-terminal domain (NTD), spanning residues 1-114, is predominantly unstructured and contains regulatory features. This region includes a conserved BRC repeat-binding motif that mediates a critical interaction with BRCA2, a key RAD51 regulator. The NTD also harbors multiple phosphorylation sites, including Thr13 and Ser14, which are essential for regulating RAD51 function and facilitating its nuclear-cytoplasmic shuttling. These post-translational modifications allow for precise modulation of RAD51 activity in response to cellular signals and DNA damage (22,23). The core domain (residues 115-339) is the most conserved and serves as the primary site for recombinase activity. It contains the Walker A and B motifs, which are essential for ATP binding and hydrolysis, as well as the structural components required for recombinase function (Fig. 2).

ATP hydrolysis provides the energy required for RAD51 filament assembly and strand exchange (24,25). Within this domain, two DNA-binding loops (L1 and L2) facilitate interactions with both single- and double-stranded DNA, aiding in homology recognition and strand invasion during homologous recombination (HR). These loops enable RAD51 to efficiently bind DNA and mediate repair processes. The core domain of RAD51 consists of two subdomains connected by a linker and adopts a RecA-like fold, a structural feature shared among recombinases across species. This conservation highlights the evolutionary significance of this fundamental step in DNA repair (26).

RAD51 primarily exists as a quaternary structure in the form of heptameric rings, which undergo conformational changes upon binding to single-stranded DNA (ssDNA). In the presence of ATP, these rings dissociate and reassemble onto ssDNA, forming active nucleoprotein filaments essential for strand invasion and homology searching. The dynamic assembly and disassembly of these filaments serve as a critical regulatory mechanism in HR (27).

PALB2

PALB2 is a larger protein, comprising 1,186 amino acids, that functions as a molecular scaffold in the HR pathway by coordinating the activities of multiple DNA repair factors. Its structure consists of several distinct domains that contribute to its broad functionality. The N-terminal coiled-coil domain (residues 9-44) interacts with BRCA1, a critical step in assembling the BRCA1-PALB2-BRCA2 complex. This interaction enables PALB2 to recruit BRCA2 and RAD51 to sites of DNA damage, underscoring its essential role in HR (19,28). Adjacent to the coiled-coil domain, the N-terminal region (residues 1-200) contains an evolutionarily conserved sequence that preferentially binds D-loop structures, which are key intermediates in HR. The C-terminal region (residues 853-1186) features a predicted DNA-binding domain with a strong affinity for both single- and double-stranded DNA. The presence of multiple DNA-binding regions suggests that PALB2 not only identifies and stabilizes DNA structures requiring repair but also serves as a critical scaffold during the repair process (29,30).

A defining feature of PALB2 is its chromatin association motif (ChAM, residues 395-446), which facilitates chromatin localization and enables the protein to respond to DNA damage (Fig. 3). This domain enables PALB2 to function as a potential sensor of chromatin state, linking DNA repair processes to broader chromatin organization. PALB2 contains an MRG15-binding domain (residues 611-764), which interacts with Mrg15, a component of histone acetyltransferase and deacetylase complexes. This association connects PALB2 to chromatin remodeling processes, where it facilitates gene silencing to support DNA repair (19,31). In the C-terminal portion (residues 853-1186), PALB2 possesses a WD40 domain that forms a seven-bladed β-propeller structure. This domain serves as the primary interaction site for BRCA2 and is also involved in DNA binding and RAD51 recruitment. Due to its ability to mediate multiple protein-protein interactions, the WD40 domain plays a key role in establishing PALB2 as a molecular scaffold essential for assembling the homologous recombination (HR) machinery at sites of DNA damage (19,29,32). Throughout its structure, PALB2 contains two RAD51-binding domains (residues 101-184 and 850-1186), which facilitate the recruitment of RAD51 to DNA lesions. This interaction strengthens the connection between PALB2 and the recombinase components essential for HR, underscoring its central role in orchestrating HR complex assembly and function (19,33).

Similar to other essential DNA repair proteins, RAD51 and PALB2 have complex modular structures that reflect their functional roles in HR and their significance in cancer susceptibility. Structural alterations in these proteins can severely affect their function and compromise genomic stability. For instance, mutations in the RAD51 core domain may impair its ATPase activity or DNA-binding ability, leading to defective HR and an increased cancer risk. Similarly, mutations in the coiled-coil domain of PALB2 may weaken its binding affinity for BRCA1, while alterations in the WD40 domain could disrupt its interaction with BRCA2, thereby impairing HR activity (32,34-36).

Molecular functions

RAD51: orchestrating homologous recombination

RAD51, a conserved recombinase, is essential for HR, ensuring genetic integrity in breast, ovarian and prostate tissues. Along with its paralogs (RAD51B, RAD51C, RAD51D, XRCC2 and XRCC3), RAD51 assembles nucleoprotein filaments on ssDNA, facilitating the search for homologous sequences and strand invasion. This process is fundamental for repairing DNA double-strand breaks (DSBs) and interstrand crosslinks (32,33,37-39).

The functional scope of RAD51 transcends its traditional role in homologous recombination repair, incorporating essential defense against oxidative damage, a major factor in genomic instability inside cancer cells. Evidence suggests that RAD51 is overexpressed in high-grade serous ovarian cancer and is associated with unfavorable prognosis. This overexpression seems functionally associated with the regulation of heightened reactive oxygen species (ROS) levels commonly seen in aggressive cancers (40). The reduction or suppression of RAD51 leads to G2/M cell cycle arrest, indicating its significance in cellular responses to oxidative stress. Mechanistically, RAD51 aids in the repair of oxidative DNA damage by participating in homology-directed repair pathways and specialized responses to oxidative stress. The protein seems to regulate mitochondrial function, affecting superoxide production and overall cellular redox balance. The protective function of RAD51 against oxidative damage may partly elucidate why its overexpression provides survival benefits to cancer cells in microenvironments marked by increased ROS (41,42).

RAD51 constructs a helical nucleoprotein filament on ssDNA, enabling the recognition and invasion of homologous DNA sequences for accurate DNA repair. Binding to ssDNA stabilizes the filament, whereas mutations that destabilize the RAD51-ssDNA complex result in deficient DSB repair (43-45). The RAD51-ssDNA filament scans neighboring DNA duplexes for homologous sequences. Upon identifying a match, RAD51 facilitates strand invasion, allowing ssDNA to penetrate the double-stranded template and form a displacement loop (D-loop). This step is critical for RAD51-dependent DNA repair, as strand exchange is a key function of the protein. The assembly and disassembly of the RAD51 filament are regulated by replication protein A (RPA) and RAD51 paralogs, which modulate filament activity and stability (46-48).

The RAD51-ssDNA filament scans adjacent DNA duplexes for homologous sequences. Upon identifying homologous regions, RAD51 facilitates strand invasion, allowing ssDNA to penetrate the double-stranded template and form a D-loop. This step is essential for accurate DNA repair and relies on the ability of RAD51 to mediate strand exchange. The assembly and disassembly of RAD51 filaments are regulated by proteins such as RPA and RAD51 paralogs, which modulate filament stability and function (46-48). The ATPase activity of RAD51 is critical for its role in DNA strand exchange. Calcium ions regulate this activity by reducing ATP hydrolysis, thereby stabilizing the active RAD51-ATP-ssDNA filament, which is necessary for efficient strand exchange (49). Differential extension of dsDNA associated with RAD51 filaments enhances homology recognition and strand exchange by accelerating the dissociation of non-homologous dsDNA, allowing for the efficient replacement with homologous sequences (50). Proteins such as RTT105 and RAD54 further enhance RAD51 function; RTT105 promotes RAD51 assembly and strand exchange, while RAD54 cooperates with RAD51 to facilitate DNA pairing and unwinding, both of which are essential for homology searching (46,51).

RAD51 paralogs and BRCA2 play a crucial role in remodeling and stabilizing RAD51 filaments, ensuring their function in HR. These proteins assist in nucleating and stabilizing RAD51 filaments on ssDNA, particularly at dsDNA-ssDNA junctions (48,52,53). Beyond its catalytic role, RAD51 has non-catalytic functions, such as preventing error-prone DNA repair mechanisms that could compromise genomic integrity. Additionally, RAD51 interacts with various other proteins, including nucleases and helicases, influencing multiple aspects of the DNA repair process (Fig. 4). Mutations in RAD51 can disrupt HR, leading to genomic instability and increasing the risk of malignancies such as breast and ovarian cancer (24,54,55).

Initiation of HRR and the interaction
mechanisms of RAD51, PALB2 and BRCA2. (A) The
mechanism of HR initiation and the role of other proteins in
facilitating this process. The repair of a double-stranded DNA
break requires several essential proteins and complexes. The MRN
complex first identifies the DSB and attaches to the location.
Subsequently, it recruits ATM kinase, which phosphorylates
BRCA1, facilitating its recruitment to the site of injury.
During the S phase, RPA protein complex binds to the exposed
single-stranded DNA created by end resection, allowing the
BRCA1-PALB2-BRCA2 complex to load RAD51 onto the DNA,
enabling HRR, where RAD51 actively searches for a homologous
DNA strand to repair the break, with RPA being displaced once
RAD51 is bound. (B) Another possible pathway is via the NHEJ
pathway. In the G1 phase of the cell cycle, NHEJ is
primarily triggered if there is a presence of protein 53BP1 which
plays a crucial role in the process, and KU70/KU80 heterodimers
binds to the broken ends, facilitating the recruitment of the
DNA-PKcs, XRCC4 and LIG4 to finally join the DNA ends back
together. (C) The effect of presence and absence of BRCA1 on
RAD51 and PALB2. In the BRCA +/+ state, all
the interactions and recruitments among the genes and proteins in
normal cell is stable. PALB2 recruit BRCA1 by direct
interaction and it also interacts with MRG15, which is a
chromodomain containing proteins that bind histone H3K36me3.
Sufficient amount of RAD51 is also loaded onto the DNA and
RNF168 acts as stabilizing factor for these interactions. Whereas
in the BRCA -/- state, the Shieldin complex is active which
inhibits the binding of BRCA2-PALB2-RAD51 complex
effectively preventing the RAD51 recruitment to the damage
site leading to impaired HRR. HRR, homologous recombination repair;
HR, homologous recombination; MRN, Mre11-Rad50-Nbs1; DSB,
double-strand break; ATM Ataxia Telangiectasia Mutated; RPA,
replication protein A; NHEJ, non-homologous end joining; DNA-PKcs,
DNA-dependent protein kinase catalytic subunit; LIG4, Ligase IV;
H3K36me3, H3 trimethylated at lysine 36.

Figure 4

Initiation of HRR and the interaction mechanisms of RAD51, PALB2 and BRCA2. (A) The mechanism of HR initiation and the role of other proteins in facilitating this process. The repair of a double-stranded DNA break requires several essential proteins and complexes. The MRN complex first identifies the DSB and attaches to the location. Subsequently, it recruits ATM kinase, which phosphorylates BRCA1, facilitating its recruitment to the site of injury. During the S phase, RPA protein complex binds to the exposed single-stranded DNA created by end resection, allowing the BRCA1-PALB2-BRCA2 complex to load RAD51 onto the DNA, enabling HRR, where RAD51 actively searches for a homologous DNA strand to repair the break, with RPA being displaced once RAD51 is bound. (B) Another possible pathway is via the NHEJ pathway. In the G1 phase of the cell cycle, NHEJ is primarily triggered if there is a presence of protein 53BP1 which plays a crucial role in the process, and KU70/KU80 heterodimers binds to the broken ends, facilitating the recruitment of the DNA-PKcs, XRCC4 and LIG4 to finally join the DNA ends back together. (C) The effect of presence and absence of BRCA1 on RAD51 and PALB2. In the BRCA +/+ state, all the interactions and recruitments among the genes and proteins in normal cell is stable. PALB2 recruit BRCA1 by direct interaction and it also interacts with MRG15, which is a chromodomain containing proteins that bind histone H3K36me3. Sufficient amount of RAD51 is also loaded onto the DNA and RNF168 acts as stabilizing factor for these interactions. Whereas in the BRCA -/- state, the Shieldin complex is active which inhibits the binding of BRCA2-PALB2-RAD51 complex effectively preventing the RAD51 recruitment to the damage site leading to impaired HRR. HRR, homologous recombination repair; HR, homologous recombination; MRN, Mre11-Rad50-Nbs1; DSB, double-strand break; ATM Ataxia Telangiectasia Mutated; RPA, replication protein A; NHEJ, non-homologous end joining; DNA-PKcs, DNA-dependent protein kinase catalytic subunit; LIG4, Ligase IV; H3K36me3, H3 trimethylated at lysine 36.

RAD51 activity is closely linked to other DNA repair pathways, including non-homologous end joining (NHEJ) and single-strand annealing (SSA). NHEJ primarily repairs DSBs when homologous templates are unavailable. However, this error-prone mechanism can promote genomic instability, particularly when RAD51 function is compromised, shifting the repair process toward non-homologous mechanisms. RAD51 prevents non-conservative repair pathways such as SSA and alternative end-joining (A-EJ) by occupying ssDNA, thereby inhibiting the annealing step required for these pathways. This function is independent of its role in promoting gene conversion (GC). Silencing or impairing RAD51 increases SSA and A-EJ activity but does not affect classical NHEJ (C-NHEJ) (56,57). Under low DSB conditions, GC is the preferred repair pathway; however, as DSB load increases, GC is suppressed while SSA becomes more prominent. This shift is not due to RAD51 availability but is influenced by additional factors such as 53BP1 and RAD52 (58). RAD51-mediated HR can inhibit NHEJ, particularly at replication fork barriers, where RAD51 acts as an early responder to stalled forks, preventing NHEJ from accessing these sites (59). Some repair mechanisms, such as single-strand template repair in gene editing, can occur independently of RAD51, instead relying on proteins such as RAD52 and RAD59 (60).

BRCA2 regulates the HR activity of RAD51 through two RAD51-binding domains: A core domain containing eight BRC repeats and a C-terminal RAD51-binding domain (CTRBD) with a phosphorylation site. The CTRBD enhances HR by stabilizing RAD51 oligomers and nucleofilaments, thereby improving HR efficiency. Mutant BRCA2 lacking RAD51-interacting mutations in the CTRBD fails to support HR when tested with the full-length protein but retains partial function when fused to an essential BRCA2 domain. Exogenous CTRBD expression promotes HR without affecting NHEJ efficiency and confers resistance to DNA-damaging treatments. This resistance depends on endogenous BRCA2, demonstrating the potential of therapeutic strategies aimed at enhancing CTRBD activity. The expression of CTRBD facilitates RAD51 foci formation, indicating efficient DNA repair. These findings suggest that peptides derived from CTRBD could serve as protective agents for normal tissues during cancer therapy or as sensitizers to enhance the efficacy of existing treatments in tumor cells (61-64).

PALB2: interaction with BRCAs and RAD51

PALB2 functions as a tumor suppressor and plays a critical role in the DNA damage response. It interacts with BRCA1 and BRCA2, forming a complex essential for recruiting and loading RAD51 onto DNA, thereby acting as a scaffold to initiate HR (20,38,65,66). The ability of PALB2 to interact with chromatin and form oligomers is necessary for assembling the BRCA2-RAD51 repair complex at sites of DNA damage, independent of other DNA damage checkpoint proteins (67,68). The N-terminal coiled-coil motif of PALB2 regulates its self-association, a process critical for its function in HR. This self-interaction competes with the PALB2-BRCA1 interaction, enabling a switch that activates HR when required (68,69). In collaboration with BRCA2, PALB2 stimulates polymerase η (Polη) in recombination-associated DNA synthesis at blocked replication forks, demonstrating its role beyond D-loop formation (70).

Additionally, PALB2 participates in the G2/M checkpoint response, linking BRCA1 and BRCA2 in checkpoint activation and maintenance to prevent chromosomal abnormalities following DNA damage (66). PALB2 also interacts with RNF168, which connects the HR machinery to histone ubiquitylation, facilitating the assembly of HR complexes at DNA breaks (71,72). Beyond its role in HR repair, PALB2 is involved in the Fanconi anemia (FA) pathway, a crucial response to interstrand DNA crosslinks. Biallelic PALB2 mutations not only disrupt HR repair but also contribute to the development of FA, a genetic disorder associated with increased cancer susceptibility, particularly breast and ovarian cancers. This link underscores the multifaceted role of PALB2 in tumorigenesis through diverse DNA repair mechanisms (20,73,74).

Synergistic interaction with other HRR genes

RAD51 and PALB2

PALB2 plays a critical role in homologous recombination by acting as a key partner of RAD51. Its primary function is to facilitate the loading of RAD51 onto ssDNA, enabling strand invasion. This interaction promotes the formation of the RAD51 filament on ssDNA, a process essential for homology searching and DNA repair. PALB2 enhances RAD51 activity and serves as a molecular bridge between BRCA1 and BRCA2, stabilizing the RAD51 filament and optimizing its recombinase function (33,38,75) (Fig. 4A and B).

BRCA1-PALB2-BRCA2-RAD51 complex

The intricate structure of this protein complex highlights the coordinated function of the HR repair pathway. BRCA1 serves as the primary sensor of DNA DSBs, facilitating the recruitment of PALB2 to the damage site. Acting as a molecular scaffold, PALB2 aids in the localization of BRCA2, which, in turn, facilitates the loading of RAD51 onto ssDNA at the break site. RAD51 then assembles into nucleoprotein filaments that catalyze the search for homologous sequences and initiate strand invasion, a defining step of HR repair (38,76,77). The interdependence of these components is evident in how each protein enhances the function of the others. The BRCA1-PALB2 interaction strengthens the ability of BRCA1 to recognize DNA damage, thereby improving the capacity of BRCA2 to load RAD51. This series of interactions results in repair efficiency that surpasses the combined effectiveness of the individual proteins. Mutations in any of these genes can severely disrupt the HR repair process, leading to genomic instability and an increased risk of malignancies (38,66,68) (Fig. 4A and B).

RAD51 paralogs (XRCC2, RAD51C)

RAD51 paralogs, such as XRCC2 and RAD51C, interact synergistically with RAD51 to enhance its function. These proteins share structural similarities with RAD51 and are essential for stabilizing RAD51 nucleoprotein filaments on ssDNA, thereby improving the efficiency of homology searching and strand exchange (25,48,57). Together with other paralogs, XRCC2 and RAD51C form distinct protein complexes that operate at different stages of the HR process. The BCDX2 complex (RAD51B-RAD51C-RAD51D-XRCC2) facilitates RAD51 binding to ssDNA, while the CX3 complex (RAD51C-XRCC3) plays a key role in resolving Holliday junctions. The division of functions among these paralogs enhances the robustness and efficiency of the HR pathway (78,79).

BRCA1-FANCN/RAD51 interaction

The interaction between BRCA1, FANCN (PALB2) and RAD51 establishes a crucial link between the FA pathway and HR repair. FANCN/PALB2 acts as a bridge between these pathways, facilitating their cooperation in specific repair scenarios, particularly in resolving DNA interstrand crosslinks (ICLs). BRCA1 interacts with FANCN/PALB2, which subsequently associates with BRCA2 and RAD51, enabling the recruitment of HR factors to sites of ICL damage processed by the FA pathway. This interaction ensures a coordinated response to complex DNA damage, highlighting the interconnected nature of DNA repair mechanisms (20,80-82).

Antagonistic interactions with other HRR genes

53BP1-RAD51 interplay

The antagonistic relationship between 53BP1 and RAD51 highlights the intricate balance among DNA repair mechanisms. While RAD51 facilitates HR, 53BP1 promotes NHEJ, an error-prone repair pathway that directly joins DNA ends without requiring extensive homology. Although NHEJ is less accurate than HR, it provides a rapid response in time-sensitive situations (58,83,84). 53BP1 competes with BRCA1 for binding at DNA damage sites, potentially inhibiting the recruitment of HR factors such as RAD51. This competition is cell cycle-dependent, with 53BP1 favoring NHEJ during the G1 phase, whereas BRCA1 counteracts 53BP1 in the S and G2 phases to facilitate HR. The interplay between these pathways ensures the selection of an appropriate repair mechanism based on the cell cycle stage and the nature of the DNA damage (85-88) (Fig. 4B).

Srs2 and RAD51

While several interactions enhance RAD51 function, certain proteins exert antagonistic effects. Srs2 acts as a major anti-recombinase by specifically targeting RAD51 filaments, promoting their disassembly and disrupting the RAD51-ssDNA filament. Srs2 interacts with RAD51 and stimulates ATP hydrolysis within the filament, thereby reducing RAD51 availability for recombination. This antagonistic activity is counteracted by the RAD55-RAD57 complex, which inhibits Srs2 and helps maintain RAD51 filament stability (89,90).

PARP Inhibitors and BRCA1/2 mutations

The use of PARP inhibitors (PARPi) in BRCA1/2-mutated cells exemplifies the concept of synthetic lethality, an approach leveraged in cancer treatment. PARPi selectively target cells with HR repair deficiencies, particularly those harboring BRCA1/2 mutations (Fig. 5). These inhibitors function by preventing the repair of single-strand breaks, which can accumulate and lead to DSBs that require HR for resolution (91-96). However, the effectiveness of these drugs may be compromised by compensatory DNA repair pathways. RAD51 paralogs, such as XRCC2, can partially restore HR functions in BRCA-mutated cells, reducing their sensitivity to PARPi. This resistance highlights the antagonistic interplay between DNA repair mechanisms and the challenges associated with targeting specific pathways for cancer treatment (91,97,98).

RAD51 and PALB2 and its association with various types of cancer

Mutations in RAD51 and its paralogs have been associated with an increased susceptibility to breast cancer. Biallelic alterations in RAD51C have been linked to Fanconi anemia, while monoallelic alterations elevate the risk of breast and ovarian cancer (99-101). Similarly, RAD51D mutations have been implicated in ovarian cancer predisposition, with RAD51C and RAD51D mutations specifically associated with an increased risk of ovarian cancer (102,103). Additionally, variants in RAD51B have been linked to both breast and ovarian cancer, further emphasizing the critical role of RAD51 in these malignancies (104). PALB2 mutations are recognized as significant risk factors for breast cancer. Studies suggest that the lifetime risk for female carriers of PALB2 mutations may be comparable to that of BRCA2 mutation carriers (105,106). Although PALB2 mutations are relatively rare, they play a notable role in hereditary breast cancer. By contrast, RAD51C mutations are less commonly observed in familial breast cancer cases (65,107) (Table I).

Table I

A summary of RAD51 and PALB2 and its association with various cancer.

Table I

A summary of RAD51 and PALB2 and its association with various cancer.

GeneAssociated diseaseSummary
RAD51Breast cancerOverexpression of RAD51 is common in breast cancer and is linked to more genomic instability, aggressive tumor behavior and a poor outcome. Changes or problems with RAD51 can make homologous recombination repair less effective. This makes tumors more vulnerable to DNA-damaging agents but also makes treatment less effective.
Ovarian cancerRAD51 dysregulation in ovarian cancer is associated with HRD, which affects the efficacy of platinum-based chemotherapy and PARP inhibitors. Elevated RAD51 expression is indicative of resistance to these therapies and correlates with poorer clinical outcomes.
Prostate cancerMutations or altered expression of RAD51 and its paralogs are associated with prostate cancer, potentially contributing to genomic instability and influencing sensitivity to DNA-damaging therapies.
Pancreatic cancerOverexpression enhances proliferation and aerobic glycolysis through HIF1α, which is associated with reduced survival rates. Homologous recombination deficiency in PALB2-mutant tumors increase sensitivity to PARP inhibitors.
PALB2Breast cancerGermline mutations in PALB2 are associated with a substantial increase in breast cancer risk, with affected individuals experiencing a lifetime risk of up to 53%. PALB2 serves as a crucial scaffold connecting BRCA1 and BRCA2; its absence disrupts homologous recombination, resulting in heightened susceptibility and affecting the response to PARP inhibitors.
Ovarian cancerPALB2 mutations are infrequent in ovarian cancer; however, they still present an elevated risk. Tumors exhibiting PALB2 loss frequently demonstrate HRD, which can be therapeutically targeted using DNA-damaging agents and PARP inhibitors.
Prostate cancerGermline mutations associated with aggressive phenotypes. Biallelic inactivation leads to homologous recombination deficiency, allowing for the stratification of patients for PARP inhibitor therapy. Somatic alterations affect the capacity for DNA repair.
Pancreatic cancerGermline mutations in PALB2 are linked to a moderately elevated risk of pancreatic cancer. The compromised DNA repair capacity in these instances may guide targeted treatment approaches.

[i] HRD, homologous recombination deficiency; PARP, poly-(ADP-ribose) polymerase; HIF1α, hypoxia-inducible factor 1α.

In ovarian cancer, RAD51 and PALB2 mutations influence not only cancer susceptibility but also treatment outcomes. Ovarian cancers with RAD51- and PALB2-deficient homologous recombination, along with other repair dysfunctions, exhibit increased sensitivity to platinum-based chemotherapy and PARPi. This sensitivity arises from synthetic lethality, wherein the simultaneous deficiency of two DNA repair pathways, HR and base excision repair, leads to cell death (Fig. 5). However, the restoration of RAD51 function has been associated with acquired resistance to these therapies, underscoring the dynamic nature of HR pathway alterations in cancer progression and treatment (108-112). Elevated RAD51 levels correlate with poor prognosis and reduced progression-free survival in ovarian cancer patients. RAD51 also serves as a predictive biomarker for platinum resistance, indicating poorer treatment outcomes (38,113) (Table I).

Germline PALB2 mutations are linked to an increased risk of prostate cancer, particularly in its aggressive forms. Genome-wide association studies have identified RAD51B, a RAD51 paralog, as a susceptibility gene for prostate cancer. Additionally, somatic alterations in RAD51 and PALB2 contribute to HRD in a subset of prostate cancers, potentially influencing treatment strategies, including the use of PARPi (38,114,115). In prostate cancer, RAD51 expression is frequently upregulated, enhancing DNA repair capacity and contributing to therapy resistance, particularly against radiation and chemotherapy. EGFR signaling regulates RAD51 expression, promoting DNA repair and epithelial-mesenchymal transition (EMT), which drive intrinsic resistance in prostate cancer cells (116). Furthermore, the Jak2-Stat5a/b signaling pathway is essential for RAD51 expression and its inhibition sensitizes prostate cancer cells to radiation by impairing HR-mediated DNA repair (117) (Table I).

PALB2 functions as a tumor suppressor and plays a critical role in homologous recombination by facilitating the recruitment of BRCA2 and RAD51 to sites of DNA damage. It enhances the recombinase activity of RAD51, which is essential for assembling the synaptic complex during HR (118,119). Mutations in PALB2 lead to HRD, increasing the susceptibility of cancer cells to DNA-damaging agents such as PARP inhibitors. In prostate cancer, biallelic PALB2 inactivation is associated with HRD and serves as a criterion for stratifying patients for PARPi therapy (119) (Table I).

In pancreatic cancer, RAD51 overexpression has been shown to promote cancer cell proliferation and regulate aerobic glycolysis by targeting hypoxia-inducible factor 1α (HIF1α). This overexpression correlates with poor survival outcomes in pancreatic cancer patients (120,121). A study identified novel germline missense variants of PALB2 (p.Ser64Leu and p.Pro104Leu) in patients with familial pancreatic cancer. These variants impair the DNA damage response by disrupting the recruitment of PALB2 and RAD51 to DNA damage foci, leading to defective homologous recombination and increased cellular sensitivity to ionizing radiation and PARP inhibitors. These findings suggest that patients harboring these PALB2 variants may benefit from personalized treatments incorporating these therapeutic agents (122) (Table I).

Mutational landscape of RAD51 and PALB2

In the RAD51 gene analysis, a total of 65,779 samples across all tissue types were examined, including 5,718 samples specifically related to breast cancer (123). Among these, 28 samples contained mutations, accounting for ~0.49% of the breast cancer cases analyzed. In the case of the PALB2 gene, 69,198 total samples were analyzed across all tissue types, with 5,856 samples from breast cancer cases (123). Of these, 153 samples exhibited mutations, representing ~2.61% of the breast cancer cases tested. Regarding gene expression in breast cancer, 1,104 expression profiles were recorded for both RAD51 and PALB2. RAD51 was overexpressed in 109 cases, while PALB2 was overexpressed in 187 cases (16.94%) and under-expressed in 7 cases (0.63%; Tables II and III).

Table II

Gene mutation analysis of RAD51 and PALB2 genes on breast, ovarian, prostate and pancreatic cancers among all other cancer types (https://cancer.sanger.ac.uk/cosmic/gene/analysis).

Table II

Gene mutation analysis of RAD51 and PALB2 genes on breast, ovarian, prostate and pancreatic cancers among all other cancer types (https://cancer.sanger.ac.uk/cosmic/gene/analysis).

GeneSample type, nMutation, nMutation percentage
RAD51All cancer types, 65,7793690.56
Breast cancer, 5,718280.49
Ovarian cancer, 1,340231.72
Prostate cancer, 3,258310.95
Pancreatic cancer, 2,656170.64
PALB2All cancer types, 69,1981,2721.84
Breast cancer, 5,8561532.61
Ovarian cancer, 1,524291.9
Prostate cancer, 3,8191423.72
Pancreatic cancer, 2,935361.23

Table III

Gene expression analysis of RAD51 and PALB2 in carcinomas such as breast, ovary, pancreas and prostate (https://cancer.sanger.ac.uk/cosmic/gene/analysis).

Table III

Gene expression analysis of RAD51 and PALB2 in carcinomas such as breast, ovary, pancreas and prostate (https://cancer.sanger.ac.uk/cosmic/gene/analysis).

Gene NameGene expression, nOverexpressed, nOverexpressed, %Underexpressed, nUnderexpressed, %
RAD51All cancers
65,779
Breast cancer
1,1041099.87--
Ovarian cancer
26662.26--
Pancreas cancer
17942.23--
Prostate cancer
498142.81--
PALB2All cancer types
69,198
Breast cancer
1,10418716.9470.63
Ovarian cancer
26672.63103.76
Pancreas cancer
17984.4731.68
Prostate cancer
498316.2271.41

For ovarian cancer, 65,779 total samples were analyzed for RAD51, including 1,340 ovarian cancer-specific samples. Among these, 23 cases (1.72%) carried mutations. In the case of PALB2, 69,198 total samples were analyzed, with 1,524 related to ovarian cancer, among which 29 cases (1.9%) harbored mutations. Regarding gene expression in ovarian cancer, 266 expression profiles were recorded, with RAD51 overexpressed in 6 cases (2.26%). PALB2 was overexpressed in seven cases (2.63%) and under-expressed in 10 cases (3.76%; Tables II and III) (123).

In prostate cancer, RAD51 mutations were detected in 0.95% (31/3,258) of samples, with overexpression observed in 2.81% (14/498) of cases. PALB2 mutations were found in 3.72% (142/3,819) of samples, with 6.22% (31/498) showing overexpression and 1.41% (7/498) showing under-expression. In pancreatic cancer, RAD51 mutations were identified in 0.64% (17/2,656) of samples, with 2.23% (4/179) exhibiting overexpression. PALB2 mutations occurred in 1.23% (36/2,935) of cases, with 4.47% (8/179) showing overexpression and 1.68% (3/179) showing under-expression (Tables II and III) (123).

Clinical significance of RAD51 and PALB2 in breast and ovarian cancer

The growing understanding of RAD51 and PALB2 mutations has markedly advanced the management of hereditary cancer syndromes. Current guidelines from the National Comprehensive Cancer Network (NCCN; https://www.nccn.org/guidelines/category_1) and the American Society of Clinical Oncology (ASCO; https://www.asco.org/search?q=genetic_testing) recommend genetic testing for individuals with specific cancer histories, including early-onset breast cancer, male breast cancer, triple-negative breast cancer and familial cancer predispositions (4,124,125). These mutations confer substantial cancer risks, with PALB2 variations associated with a 2- to 4-fold increase in breast cancer risk (33-58% lifetime risk) (19,123), whereas RAD51 variants have been linked to increased risks of breast, ovarian, pancreatic and prostate cancers (18,100,110,122,126,127). Genetic test results range from pathogenic to benign, necessitating expert molecular genetic interpretation and genetic counseling (128,129). Management strategies for mutation carriers include enhanced surveillance protocols such as early mammography, ultrasound, MRI and targeted screenings (130). Preventive surgeries, including risk-reducing mastectomy or risk-reducing salpingo-oophorectomy, can reduce the risk of certain cancers by >90% (131-133). Additionally, chemoprevention strategies using selective estrogen receptor modulators such as tamoxifen and raloxifene, or aromatase inhibitors, are being explored as preventive measures (134-136). Critically, these genetic insights have revolutionized cancer treatment, particularly through targeted therapies such as PARP inhibitors, which exploit synthetic lethality in homologous recombination-deficient cancers (Fig. 5) (137). Personalized chemotherapy selection, including platinum-based regimens, has further refined treatment approaches (138). Investigational therapies, such as ATR inhibitors (139,140) and WEE1 inhibitors (141), hold promise for expanding treatment options. Ultimately, these advances contribute to more precise and individualized cancer prevention and treatment strategies, improving patient outcomes.

PARP inhibitors play a crucial role in the treatment of breast and ovarian malignancies, particularly in individuals with BRCA mutations or HRD. These drugs leverage synthetic lethality by inhibiting two DNA repair pathways, inducing cytotoxicity in cancer cells while sparing normal cells (142). They have demonstrated significant efficacy in improving progression-free survival, especially in BRCA-mutated cancers (143). PARPi have shown potential beyond BRCA-mutated cancers, offering benefits to patients with additional HRR pathway deficiencies or platinum-resistant tumors (144). Efforts to enhance their efficacy have led to combination therapies involving chemotherapy, antiangiogenic agents and immunotherapy (145).

Several PARPi have been approved or are in late-stage clinical trials for breast and ovarian cancer treatment. Olaparib has demonstrated efficacy as a monotherapy in BRCA-related tumors in phase II trials (146). Talazoparib was approved by the FDA in October 2018 for treating metastatic germline BRCA1/2-mutated breast cancer. Rucaparib and niraparib have been approved for ovarian cancer treatment (147). Veliparib is in late-stage clinical development and has been investigated in combination with chemotherapy agents such as carboplatin, paclitaxel and temozolomide (145). These inhibitors have demonstrated efficacy across various clinical settings, including neoadjuvant, adjuvant and metastatic treatments for breast and ovarian cancer (148). Numerous clinical trials have assessed the efficacy and safety of PARPi across different cancer types: SOLO-1 trial (NCT01844986) evaluated Olaparib as a maintenance treatment for newly diagnosed advanced ovarian cancer with BRCA mutations. This phase III trial showed a significant improvement in progression-free survival compared with placebo (149). OlympiA trial (NCT02032823) is investigating Olaparib in the adjuvant setting for high-risk HER2-negative breast cancer patients with BRCA1/2 mutations, including potential cases with RAD51 and PALB2 mutations (150). TAPUR trial (NCT02693535) is exploring Olaparib in various solid tumors, including those with mutations in HRR genes such as RAD51 and PALB2 (151). EMBRACA trial (NCT01945775) examined Talazoparib in patients with advanced breast cancer and germline BRCA mutations. This phase III study demonstrated superior progression-free survival compared with chemotherapy (152). ARIEL2 trial (NCT01891344) assessed Rucaparib in patients with platinum-sensitive, high-grade ovarian cancer. This phase II trial developed a tumor genomic profiling assay to quantify HR loss of heterozygosity using next-generation sequencing (153). Phase III trial (NCT02470585) evaluated Veliparib in combination with platinum-based chemotherapy for locally advanced or metastatic breast cancer, yielding favorable outcomes (152). PRIMA study (NCT02655016) investigated Niraparib as a maintenance therapy for newly diagnosed advanced ovarian cancer. This phase III trial demonstrated improved progression-free survival across multiple biomarker-defined subgroups (154). These trials have revealed varying efficacy and safety profiles among different PARPi. For instance, Niraparib has been associated with a higher incidence of grade ≥3 adverse events compared with other PARP inhibitors (155). Expanding the indications for PARPi beyond BRCA mutations could benefit a broader patient population with DNA repair deficiencies (Fig. 6).

Therapeutic implications

For PALB2-mutated tumors lacking HR, treatment options extend beyond BRCA1/2 mutations due to their heightened sensitivity to PARP inhibitors (155-157). Similarly, RAD51 overexpression contributes to resistance against neoadjuvant endocrine therapy in estrogen receptor (ER)-positive breast carcinoma. This therapy resistance, associated with poor prognosis, is partly due to inadequate BRCA2 methylation, which fails to suppress RAD51 expression (156,158). Inhibiting RAD51 may enhance the efficacy of DNA-damaging agents and help overcome chemoresistance. RAD51 inhibitors could improve chemotherapy effectiveness while preserving genomic integrity and immune function (55,157). As a key regulator of HRR, RAD51 plays a crucial role in therapeutic resistance and cancer progression. Depletion of damage-specific DNA binding protein 2 (DDB2) leads to RAD51 destabilization, rendering triple-negative breast cancer cells more sensitive to PARPi due to compromised HR repair (157). Polymorphisms in RAD51 and XRCC3 have been associated with an increased risk of breast cancer and poor radiotherapy outcomes, highlighting the role of genetic variability in treatment response (159,160).

Pharmacologically targeting RAD51 has been shown to reduce B02-induced clonogenic survival and increase prostate cancer radioresistance. Conversely, RAD51 activators, such as RS-1, exploit RAD51 overexpression to induce synthetic lethality in cancer cells (116,119). The role of PALB2 in enhancing RAD51 activity suggests that targeting PALB2 or its interactions could serve as a viable therapeutic strategy. Disrupting BRCA1-independent mechanisms of PALB2 localization may improve treatment outcomes in BRCA1-mutant tumors that have regained HR function and developed resistance to PARPi (33). Combining RAD51 or PALB2 inhibitors with other therapies, such as radiation or PARPi, could enhance treatment efficacy. For instance, inhibiting Jak2-Stat5a/b signaling reduces RAD51 expression and sensitizes prostate cancer cells to radiation while sparing surrounding healthy tissue, making it a potential adjuvant therapy (118). RAD51 is also emerging as a potential biomarker and therapeutic target in pancreatic cancer. Inhibiting RAD51 may increase cancer cell susceptibility to DNA-damaging agents, including ionizing radiation and PARPi, by impairing their DNA repair capacity (121,160). A novel RAD51 inhibitor, CYT-0851, has demonstrated promising anticancer activity in preclinical models of pancreatic cancer, leading to significant tumor growth inhibition and, in some cases, tumor regression (161). Patients with PALB2 mutations may be particularly responsive to DNA-damaging therapies, such as ionizing radiation and PARPi, due to their impaired DNA repair capabilities (120). Similarly, targeting RAD51 with inhibitors such as CYT-0851 could enhance the efficacy of existing treatments and help overcome drug resistance in pancreatic cancer (161).

Commercial insights

The understanding of RAD51 and PALB2 functions presents several commercial potentials, particularly in oncology, where they act as therapeutic targets. Commercial tests that look for changes in PALB2 and RAD51 may help find individuals early who are more likely to get breast and ovarian cancer, allowing for proactive treatment plans. Myriad Genetics (https://myriad.com/gene-table/) provides genetic testing panels encompassing BRCA1, BRCA2 and additional genes associated with hereditary cancer, such as RAD51 and PALB2 (162). RAD51 and PALB2 are still experimental biomarkers in clinical decision-making, with minimal incorporation into FDA-approved instruments. RAD51 and PALB2 are essential elements of the HRR pathway and their modifications, such as mutations, loss of heterozygosity (LOH), or diminished RAD51 foci formation, can signify HRD, which correlates with susceptibility to PARPi and platinum-based treatments. HRD scores, which integrate genomic instability indicators such as LOH, telomeric allelic imbalance (TAI) and large-scale state transitions (LST), are used to detect HRD-positive cancers, exemplified by FDA-approved assessments such as Myriad Genetics' myChoice® CDx. This test computes a Genomic Instability Score using LOH, TAI and LST to inform PARPi therapy in ovarian cancer, although it does not directly evaluate RAD51 or PALB2 functionality. PALB2 mutations are incorporated in several next-generation sequencing (NGS) panels, such as Myriad's myRisk® and Tempus xT, which emphasize germline or somatic mutations instead of functional testing (163-166). PALB2 mutations are infrequently incorporated into expanded germline panels (such as Invitae Multi-Cancer Panel) and are not typically evaluated in regular HRD testing due to their low prevalence (~2-3%) and difficulties in variant interpretation (165,167). The therapeutic value of RAD51 focuses on functional tests such as RAD51 foci measurement, which assesses homologous recombination repair proficiency in real time. Preclinical and early-phase trials indicate that RAD51-Low scores (≤10% foci-positive cells) accurately predict sensitivity to platinum and PARPi in ovarian and triple-negative breast malignancies (168-170). Nonetheless, these assays do not possess FDA approval and encounter challenges in standardization, especially in formalin fixed and paraffin embedded samples, where preanalytical factors and scoring thresholds (such as 10 vs. 20% cutoff) influence reproducibility (167,170). Conversely, genomic scar-based HRD diagnostics such as myChoice® prevail in clinical use, although their incapacity to identify dynamic HR restoration (such as BRCA1/2 reversion mutations) (170). Current experiments (such as MITO16A/MaNGO-OV2) are assessing RAD51 foci in conjunction with genomic scores; nevertheless, widespread implementation is contingent upon standardized methodologies and validation in prospective cohorts (163,167). Consequently, whereas PALB2 is progressively incorporated into NGS panels for therapeutic selection and RAD51 shows promise as a functional biomarker, their regular clinical application necessitates further validation and incorporation into standardized HRD testing protocols (165,166).

Limitations

Notwithstanding the therapeutic promise of RAD51 and PALB2 as biomarkers, numerous obstacles impede their clinical application. The low mutation frequency of PALB2 (1-3% in hereditary breast cancers) restricts cost-effectiveness in population-wide screening, hence requiring tailored testing in high-risk groups (171). Second, ~50% of PALB2 and RAD51 variations are categorized as variants of unknown significance (VUS) owing to insufficient functional data, hence confounding risk stratification and therapeutic decision-making (172,173). Functional assays, including RAD51 foci quantification, encounter standardization challenges, especially in archival formalin fixed and paraffin embedded samples, where preanalytical factors (such as fixation duration) and laboratory HRD scoring thresholds (such as <20% RAD51-positive cells) affect reproducibility (98,163,172). Moreover, HRD genomic scar assays, although indicative of PARP inhibitor efficacy, may not adequately reflect the dynamic restoration of homologous recombination through BRCA1/2 reversion mutations or epigenetic modifications. Future methods must emphasize the integration of multigene HRD panels (such as RAD51C/D, PALB2) with functional assays to clarify VUS and evaluate real-time HR proficiency. High-throughput clustered regularly interspaced short palindromic repeats-mediated mutagenesis and AI-based categorization systems may expedite the annotation of VUS. Overcoming these constraints necessitates joint endeavors to integrate mechanistic insights with scalable diagnostic tools, hence enhancing tailored therapy methods for HRD-associated malignancies (98,163,174,175).

Conclusion

The present review highlighted the critical roles of RAD51 and PALB2 in HRR-mediated genomic stability and their significant effect on breast and ovarian cancer pathogenesis. Mutations in these genes substantially increase cancer risk, underscoring their importance in genetic testing protocols for early risk stratification. The clinical effectiveness of PARPi in HRR-deficient tumors further emphasizes the need to identify RAD51 and PALB2 mutations to inform personalized treatment strategies. However, resistance mechanisms present a major challenge, necessitating alternative therapeutic approaches, such as RAD51 inhibition or combinatorial treatment modalities.

Future research should focus on improving functional assays to assess HRR status and expanding clinical trials to include RAD51- and PALB2-mutated patient groups. These efforts, combined with the development of tailored therapies, are expected to refine precision oncology strategies and improve outcomes for patients with HRR-deficient cancers. The evolving understanding of RAD51 and PALB2 not only deepens insights into cancer biology but also paves the way for transformative therapeutic advancements.

Availability of data and materials

Not applicable.

Authors' contributions

Conceptualization was by MN and MK. Data curation was by SA. Formal Analysis was by ME-T. Initial screening and content curation was by AK. Methodology was by TG and AA. Supervision was by KA. SG wrote the original draft. Writing, reviewing and editing was by MN, SR and FA. Data authentication is not applicable. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

Funding

This research has been funded by Scientific Research Deanship at University of Ha'il-Saudi Arabia through project number (RG-24 173).

References

1 

Weinberg RA: How cancer arises. Sci Am. 275:62–70. 1996. View Article : Google Scholar

2 

Wooster R and Weber BL: Breast and ovarian cancer. N Engl J Med. 348:2339–2347. 2003. View Article : Google Scholar : PubMed/NCBI

3 

International Agency for Research on Cancer: Cancer Tomorrow. Dataviz. https://gco.iarc.fr/tomorrow/en/dataviz.

4 

Petrucelli N, Daly MB and Pal T: BRCA1- and BRCA2-Associated Hereditary Breast and Ovarian Cancer. GeneReviews® [Internet]. Adam MP, Feldman J, Mirzaa GM, et al: University of Washington; Seattle, WA: 1993-2025, https://www.ncbi.nlm.nih.gov/books/NBK1247/.

5 

Menendez JA, Folguera-Blasco N, Cuyàs E, Fernández-Arroyo S, Joven J and Alarcón T: Accelerated geroncogenesis in hereditary breast-ovarian cancer syndrome. Oncotarget. 7:11959–11971. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Fantone S, Marzioni D and Tossetta G: NRF2/KEAP1 signaling inhibitors in gynecologic cancers. Expert Rev Anticancer Ther. 24:1191–1194. 2024. View Article : Google Scholar

7 

Pokhriyal R, Hariprasad R, Kumar L and Hariprasad G: Chemotherapy resistance in advanced ovarian cancer patients. Biomark Cancer. 11:1179299X198608152019. View Article : Google Scholar :

8 

Akter S, Rahman MA, Hasan MN, Akhter H, Noor P, Islam R, Shin Y, Rahman MDH, Gazi MS, Huda MN, et al: Recent advances in ovarian cancer: Therapeutic strategies, potential biomarkers and technological improvements. Cells. 11:6502022. View Article : Google Scholar

9 

Xiong N, Wu H and Yu Z: Advancements and challenges in triple-negative breast cancer: A comprehensive review of therapeutic and diagnostic strategies. Front Oncol. 14:14054912024. View Article : Google Scholar : PubMed/NCBI

10 

Yin L, Duan JJ, Bian XW and Yu SC: Triple-negative breast cancer molecular subtyping and treatment progress. Breast Cancer Res. 22:612020. View Article : Google Scholar

11 

Campagna R, Pozzi V, Giorgini S, Morichetti D, Goteri G, Sartini D, Serritelli EN and Emanuelli M: Paraoxonase-2 is upregulated in triple negative breast cancer and contributes to tumor progression and chemoresistance. Hum Cell. 36:1108–1119. 2023. View Article : Google Scholar

12 

Le HP, Heyer WD and Liu J: Guardians of the Genome: BRCA2 and its partners. Genes (Basel). 12:12292021. View Article : Google Scholar

13 

Angeli D, Salvi S and Tedaldi G: Genetic predisposition to breast and ovarian cancers: How many and which genes to test? Int J Mol Sci. 21:11282020. View Article : Google Scholar : PubMed/NCBI

14 

Lux MP, Fasching PA and Beckmann MW: Hereditary breast and ovarian cancer: Review and future perspectives. J Mol Med (Berl). 84:16–28. 2006. View Article : Google Scholar

15 

Mekonnen N, Yang H and Shin YK: Homologous recombination deficiency in ovarian, breast, colorectal, pancreatic, non-small cell lung and prostate cancers and the mechanisms of resistance to PARP inhibitors. Front Oncol. 12:8806432022. View Article : Google Scholar

16 

Krejci L, Altmannova V, Spirek M and Zhao X: Homologous recombination and its regulation. Nucleic Acids Res. 40:5795–5818. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Grundy MK, Buckanovich RJ and Bernstein KA: Regulation and pharmacological targeting of RAD51 in cancer. NAR Cancer. 2:zcaa0242020. View Article : Google Scholar : PubMed/NCBI

18 

Nowacka-Zawisza M, Wiśnik E, Wasilewski A, Skowrońska M, Forma E, Bryś M, Różański W and Krajewska WM: Polymorphisms of homologous recombination RAD51, RAD51B, XRCC2 and XRCC3 genes and the risk of prostate cancer. Anal Cell Pathol (Amst). 2015:8286462015.

19 

Wu S, Zhou J, Zhang K, Chen H, Luo M, Lu Y, Sun Y and Chen Y: Molecular mechanisms of PALB2 function and its role in breast cancer management. Front Oncol. 10:3012020. View Article : Google Scholar

20 

Nepomuceno TC, De Gregoriis G, de Oliveira FMB, Suarez-Kurtz G, Monteiro AN and Carvalho MA: The role of PALB2 in the DNA damage response and cancer predisposition. Int J Mol Sci. 18:18862017. View Article : Google Scholar : PubMed/NCBI

21 

Yu X, Jacobs SA, West SC, Ogawa T and Egelman EH: Domain structure and dynamics in the helical filaments formed by RecA and RAD51 on DNA. Proc Natl Acad Sci USA. 98:8419–8424. 2001. View Article : Google Scholar :

22 

Subramanyam S, Ismail M, Bhattacharya I and Spies M: Tyrosine phosphorylation stimulates activity of human RAD51 recombinase through altered nucleoprotein filament dynamics. Proc Natl Acad Sci USA. 113:E6045–E6054. 2016. View Article : Google Scholar :

23 

Aihara H, Ito Y, Kurumizaka H, Yokoyama S and Shibata T: The N-terminal domain of the human RAD51 protein binds DNA: Structure and a DNA binding surface as revealed by NMR. J Mol Biol. 290:495–504. 1999. View Article : Google Scholar

24 

Thomas M, Dubacq C, Rabut E, Lopez BS and Guirouilh-Barbat J: Noncanonical roles of RAD51. Cells. 12:11692022. View Article : Google Scholar

25 

Wiese C, Hinz JM, Tebbs RS, Nham PB, Urbin SS, Collins DW, Thompson LH and Schild D: Disparate requirements for the Walker A and B ATPase motifs of human RAD51D in homologous recombination. Nucleic Acids Res. 34:2833–2843. 2006. View Article : Google Scholar

26 

Elbakry A and Löbrich M: Homologous recombination subpathways: A tangle to resolve. Front Genet. 12:7238472021. View Article : Google Scholar : PubMed/NCBI

27 

Conway AB, Lynch TW, Zhang Y, Fortin GS, Fung CW, Symington LS and Rice PA: Crystal structure of a RAD51 filament. Nat Struct Mol Biol. 11:791–796. 2004. View Article : Google Scholar

28 

Kyriukha Y, Watkins MB, Redington JM, Dastvan R, Uversky VN, Hopkins J, Pozzi N and Korolev S: The PALB2 DNA-binding domain is an intrinsically disordered recombinase. Res Sq [Preprint]. rs.3.rs-3235465. 2023.PubMed/NCBI

29 

Park Y, Zhang F and Andreassen PR: PALB2: The hub of a network of tumor suppressors involved in DNA damage responses. Biochim Biophys Acta. 1846:263–275. 2014.PubMed/NCBI

30 

Sun Y, McCorvie TJ, Yates LA and Zhang X: Structural basis of homologous recombination. Cell Mol Life Sci. 77:3–18. 2020. View Article : Google Scholar

31 

Bleuyard JY, Buisson R, Masson JY and Esashi F: ChAM, a novel motif that mediates PALB2 intrinsic chromatin binding and facilitates DNA repair. EMBO Rep. 13:135–141. 2012. View Article : Google Scholar

32 

Park JY, Singh TR, Nassar N, Zhang F, Freund M, Hanenberg H, Meetei AR and Andreassen PR: Breast cancer-associated missense mutants of the PALB2 WD40 domain, which directly binds RAD51C, RAD51 and BRCA2, disrupt DNA repair. Oncogene. 33:4803–4812. 2014. View Article : Google Scholar :

33 

Dray E, Etchin J, Wiese C, Saro D, Williams GJ, Hammel M, Yu X, Galkin VE, Liu D, Tsai MS, et al: Enhancement of the RAD51 recombinase activity by the tumor suppressor PALB2. Nat Struct Mol Biol. 17:1255–1259. 2010. View Article : Google Scholar

34 

Matos-Rodrigues G, Guirouilh-Barbat J, Martini E and Lopez BS: Homologous recombination, cancer and the 'RAD51 paradox'. NAR Cancer. 3:zcab0162021. View Article : Google Scholar

35 

Oliver AW, Swift S, Lord CJ, Ashworth A and Pearl LH: Structural basis for recruitment of BRCA2 by PALB2. EMBO Rep. 10:990–996. 2009. View Article : Google Scholar :

36 

Modesti M, Budzowska M, Baldeyron C, Demmers JA, Ghirlando R and Kanaar R: RAD51AP1 is a structure-specific DNA binding protein that stimulates joint molecule formation during RAD51-mediated homologous recombination. Mol Cell. 28:468–481. 2007. View Article : Google Scholar

37 

Lang SH, Swift SL, White H, Misso K, Kleijnen J and Quek RG: A systematic review of the prevalence of DNA damage response gene mutations in prostate cancer. Int J Oncol. 55:597–616. 2019.

38 

Foo TK and Xia B: BRCA1-dependent and independent recruitment of PALB2-BRCA2-RAD51 in the DNA damage response and cancer. Cancer Res. 82:3191–3197. 2022. View Article : Google Scholar

39 

Bonilla B, Hengel SR, Grundy MK and Bernstein KA: RAD51 gene family structure and function. Annu Rev Genet. 54:25–46. 2020. View Article : Google Scholar :

40 

Wang Z, Jia R, Wang L, Yang Q, Hu X, Fu Q, Zhang X, Li W and Ren Y: The emerging roles of RAD51 in cancer and its potential as a therapeutic target. Front Oncol. 12:9355932022. View Article : Google Scholar

41 

Fantone S, Tossetta G, Cianfruglia L, Frontini A, Armeni T, Procopio AD, Pugnaloni A, Gualtieri AF and Marzioni D: Mechanisms of action of mineral fibres in a placental syncytiotrophoblast model: An in vitro toxicology study. Chem Biol Interact. 390:1108952024. View Article : Google Scholar

42 

Smolarz B, Michalska MM, Samulak D, Romanowicz H and Wójcik L: Polymorphism of DNA repair genes in breast cancer. Oncotarget. 10:527–535. 2019. View Article : Google Scholar

43 

Ma É, Maloisel L, Le Falher L, Guérois R and Coïc É: Rad52 oligomeric N-terminal domain stabilizes RAD51 nucleoprotein filaments and contributes to their protection against Srs2. Cells. 10:14672021. View Article : Google Scholar : PubMed/NCBI

44 

Carver A and Zhang X: RAD51 filament dynamics and its antagonistic modulators. Semin Cell Dev Biol. 113:3–13. 2021. View Article : Google Scholar

45 

Andriuskevicius T, Dubenko A and Makovets S: The inability to disassemble RAD51 nucleoprotein filaments leads to aberrant mitosis and cell death. Biomedicines. 11:14502023. View Article : Google Scholar

46 

Wang X, Zhao X, Yu Z, Fan T, Guo Y, Liang J, Wang Y, Zhan J, Chen G, Zhou C, et al: Rtt105 stimulates RAD51-ssDNA assembly and orchestrates RAD51 and RPA actions to promote homologous recombination repair. Proc Natl Acad Sci USA. 121:e24022621212024. View Article : Google Scholar : PubMed/NCBI

47 

Ma CJ, Gibb B, Kwon Y, Sung P and Greene EC: Protein dynamics of human RPA and RAD51 on ssDNA during assembly and disassembly of the RAD51 filament. Nucleic Acids Res. 45:749–761. 2017. View Article : Google Scholar :

48 

Taylor MRG, Špírek M, Chaurasiya KR, Ward JD, Carzaniga R, Yu X, Egelman EH, Collinson LM, Rueda D, Krejci L and Boulton SJ: RAD51 paralogs remodel pre-synaptic RAD51 filaments to stimulate homologous recombination. Cell. 162:271–286. 2015. View Article : Google Scholar :

49 

Bugreev DV and Mazin AV: Ca2+ activates human homologous recombination protein RAD51 by modulating its ATPase activity. Proc Natl Acad Sci USA. 101:9988–9993. 2004. View Article : Google Scholar

50 

Danilowicz C, Peacock-Villada A, Vlassakis J, Facon A, Feinstein E, Kleckner N and Prentiss M: The differential extension in dsDNA bound to RAD51 filaments may play important roles in homology recognition and strand exchange. Nucleic Acids Res. 42:526–533. 2014. View Article : Google Scholar

51 

Mazin AV, Bornarth CJ, Solinger JA, Heyer WD and Kowalczykowski SC: Rad54 protein is targeted to pairing loci by the RAD51 nucleoprotein filament. Mol Cell. 6:583–592. 2000. View Article : Google Scholar : PubMed/NCBI

52 

Akita M, Girvan P, Špírek M, Novacek J, Rueda D, Prokop Z and Krejci L: Mechanism of BCDX2-mediated RAD51 nucleation on short ssDNA stretches and fork DNA. Nucleic Acids Res. 52:11738–11752. 2024. View Article : Google Scholar : PubMed/NCBI

53 

Yang H, Li Q, Fan J, Holloman W and Pavletich N: The BRCA2 homologue Brh2 nucleates RAD51 filament formation at a dsDNA-ssDNA junction. Nature. 433:653–657. 2005. View Article : Google Scholar

54 

Richardson C: RAD51, genomic stability and tumorigenesis. Cancer Lett. 218:127–139. 2005. View Article : Google Scholar

55 

Liao C, Talluri S, Zhao J, Mu S, Kumar S, Shi J, Buon L, Munshi NC and Shammas MA: RAD51 is implicated in DNA damage, chemoresistance and immune dysregulation in solid tumors. Cancers (Basel). 14:56972022. View Article : Google Scholar : PubMed/NCBI

56 

So A, Dardillac E, Muhammad A, Chailleux C, Sesma-Sanz L, Ragu S, Le Cam E, Canitrot Y, Masson JY, Dupaigne P, et al: RAD51 protects against nonconservative DNA double-strand break repair through a nonenzymatic function. Nucleic Acids Res. 50:2651–2666. 2022. View Article : Google Scholar

57 

So A, Muhammad A, Chailleux C, Sanz L, Ragu S, Cam L, Canitrot Y, Masson J, Dupaigne P, Lopez B and Guirouilh-Barbat J: Mammalian RAD51 prevents non-conservative alternative end-joining and single strand annealing through non-catalytic mechanisms. bioRxiv. https://doi.org/10.1101/768887.

58 

Mladenov E, Staudt C, Soni A, Murmann-Konda T, Siemann-Loekes M and Iliakis G: Strong suppression of gene conversion with increasing DNA double-strand break load delimited by 53BP1 and RAD52. Nucleic Acids Res. 48:1905–1924. 2019. View Article : Google Scholar : PubMed/NCBI

59 

Willis NA, Panday A, Duffey EE and Scully R: RAD51 recruitment and exclusion of non-homologous end joining during homologous recombination at a Tus/Ter mammalian replication fork barrier. PLoS Genet. 14:e10074862018. View Article : Google Scholar :

60 

Gallagher DN, Pham N, Tsai AM, Janto AV, Choi J, Ira G and Haber JE: A RAD51-independent pathway promotes single-strand template repair in gene editing. PLoS Genet. 16:e10086892020. View Article : Google Scholar :

61 

Zhu Z, Kitano T, Morimatsu M, Tanaka A, Morioka R, Lin X, Orino K and Yoshikawa Y: BRCA2 C-Terminal RAD51-binding domain confers resistance to DNA-damaging agents. Int J Mol Sci. 23:40602022. View Article : Google Scholar :

62 

Carreira A and Kowalczykowski SC: Two classes of BRC repeats in BRCA2 promote RAD51 nucleoprotein filament function by distinct mechanisms. Proc Natl Acad Sci USA. 108:10448–10453. 2011. View Article : Google Scholar

63 

Andreassen PR, Seo J, Wiek C and Hanenberg H: Understanding BRCA2 function as a tumor suppressor based on domain-specific activities in DNA damage responses. Genes (Basel). 12:10342021. View Article : Google Scholar : PubMed/NCBI

64 

Sadeghi F, Asgari M, Matloubi M, Ranjbar M, Karkhaneh Yousefi N, Azari T and Zaki-Dizaji M: Molecular contribution of BRCA1 and BRCA2 to genome instability in breast cancer patients: Review of radiosensitivity assays. Biol Proced Online. 22:232020. View Article : Google Scholar

65 

Zhang F, Ma J, Wu J, Ye L, Cai H, Xia B and Yu X: PALB2 links BRCA1 and BRCA2 in the DNA-damage response. Curr Biol. 19:524–529. 2009. View Article : Google Scholar : PubMed/NCBI

66 

Simhadri S, Vincelli G, Huo Y, Misenko S, Foo T, Ahlskog J, Sørensen C, Oakley G, Ganesan S, Bunting S and Xia B: PALB2 connects BRCA1 and BRCA2 in the G2/M checkpoint response. Oncogene. 38:1585–1596. 2018. View Article : Google Scholar

67 

Sy S, Huen M, Zhu Y and Chen J: PALB2 regulates recombinational repair through chromatin association and oligomerization. J Biol Chem. 284:18302–18310. 2009. View Article : Google Scholar : PubMed/NCBI

68 

Song F, Li M, Liu G, Swapna GVT, Daigham NS, Xia B, Montelione GT and Bunting SF: Antiparallel coiled-coil interactions mediate the homodimerization of the DNA damage-repair protein PALB2. Biochemistry. 57:6581–6591. 2018. View Article : Google Scholar

69 

Buisson R and Masson JY: PALB2 self-interaction controls homologous recombination. Nucleic Acids Res. 40:10312–10323. 2012. View Article : Google Scholar

70 

Buisson R, Niraj J, Pauty J, Maity R, Zhao W, Coulombe Y, Sung P and Masson J: Breast cancer proteins PALB2 and BRCA2 stimulate polymerase η in recombination-associated DNA synthesis at blocked replication forks. Cell Rep. 6:553–564. 2014. View Article : Google Scholar

71 

Luijsterburg MS, Typas D, Caron MC, Wiegant WW, Van Den Heuvel D, Boonen RA, Couturier AM, Mullenders LH, Masson JY and Van Attikum H: A PALB2-interacting domain in RNF168 couples homologous recombination to DNA break-induced chromatin ubiquitylation. ELife. 6:e209222017. View Article : Google Scholar :

72 

Krais JJ, Wang Y, Patel P, Basu J, Bernhardy AJ and Johnson N: RNF168-mediated localization of BARD1 recruits the BRCA1-PALB2 complex to DNA damage. Nat Commun. 12:50162021. View Article : Google Scholar : PubMed/NCBI

73 

Pauty J, Rodrigue A, Couturier A, Buisson R and Masson JY: Exploring the roles of PALB2 at the crossroads of DNA repair and cancer. Biochem J. 460:331–342. 2014. View Article : Google Scholar : PubMed/NCBI

74 

Ducy M, Sesma-Sanz L, Guitton-Sert L, Lashgari A, Gao Y, Brahiti N, Rodrigue A, Margaillan G, Caron MC, Côté J, et al: The tumor suppressor PALB2: Inside out. Trends Biochem Sci. 44:226–240. 2019. View Article : Google Scholar : PubMed/NCBI

75 

Uemura M, Ochiai K, Morimatsu M, Michishita M, Onozawa E, Azakami D, Uno Y, Yoshikawa Y, Sasaki T, Watanabe M and Omi T: The canine RAD51 mutation leads to the attenuation of interaction with PALB2. Vet Comp Oncol. 18:247–255. 2020. View Article : Google Scholar

76 

Prakash R, Zhang Y, Feng W and Jasin M: Homologous recombination and human health: The roles of BRCA1, BRCA2 and associated proteins. Cold Spring Harb Perspect Biol. 7:a0166002015. View Article : Google Scholar

77 

Zhao W, Steinfeld JB, Liang F, Chen X, Maranon DG, Jian Ma C, Kwon Y, Rao T, Wang W, Sheng C, et al: BRCA1-BARD1 promotes RAD51-mediated homologous DNA pairing. Nature. 550:360–365. 2017. View Article : Google Scholar

78 

Saxena S, Dixit S, Somyajit K and Nagaraju G: ATR signaling uncouples the role of RAD51 paralogs in homologous recombination and replication stress response. Cell Rep. 29:551–559.e4. 2019. View Article : Google Scholar : PubMed/NCBI

79 

Berti M, Teloni F, Mijic S, Ursich S, Fuchs J, Palumbieri MD, Krietsch J, Schmid JA, Garcin EB, Gon S, et al: Sequential role of RAD51 paralog complexes in replication fork remodeling and restart. Nat Commun. 11:35312020. View Article : Google Scholar

80 

Hanenberg H and Andreassen PR: PALB2 (partner and localizer of BRCA2). Atlas Genet Cytogenet Oncol Haematol. 22:484–490. 2018.PubMed/NCBI

81 

Michl J, Zimmer J and Tarsounas M: Interplay between Fanconi anemia and homologous recombination pathways in genome integrity. EMBO J. 35:909–923. 2016. View Article : Google Scholar :

82 

Park D, Bergin SM, Jones D, Ru P, Koivisto CS, Jeon YJ, Sizemore GM, Kladney RD, Hadjis A, Shakya R and Ludwig T: Ablation of the BRCA1-PALB2 interaction phenocopies fanconi anemia in mice. Cancer Res. 80:4172–4184. 2020. View Article : Google Scholar

83 

Schwarz B, Friedl AA, Girst S, Dollinger G and Reindl J: Nanoscopic analysis of 53BP1, BRCA1 and RAD51 reveals new insights in temporal progression of DNA-repair and pathway choice. Mutat Res. 816-818:1116752019. View Article : Google Scholar : PubMed/NCBI

84 

Isono M, Niimi A, Oike T, Hagiwara Y, Sato H, Sekine R, Yoshida Y, Isobe SY, Obuse C, Nishi R, et al: BRCA1 directs the repair pathway to homologous recombination by promoting 53BP1 dephosphorylation. Cell Rep. 18:520–532. 2017. View Article : Google Scholar

85 

Malewicz M: The role of 53BP1 protein in homology-directed DNA repair: Things get a bit complicated. Cell Death Differ. 23:1902–1903. 2016. View Article : Google Scholar

86 

Ochs F, Somyajit K, Altmeyer M, Rask MB, Lukas J and Lukas C: 53BP1 fosters fidelity of homology-directed DNA repair. Nat Struct Mol Biol. 23:714–721. 2016. View Article : Google Scholar

87 

Swift ML, Beishline K, Flashner S and Azizkhan-Clifford J: DSB repair pathway choice is regulated by recruitment of 53BP1 through cell cycle-dependent regulation of Sp1. Cell Rep. 34:1088402021. View Article : Google Scholar

88 

Mohseni-Salehi FS, Zare-Mirakabad F, Sadeghi M and Ghafouri-Fard S: A stochastic model of DNA double-strand breaks repair throughout the cell cycle. Bull Math Biol. 82:112020. View Article : Google Scholar

89 

Roy U and Greene EC: The role of the Rad55-Rad57 complex in DNA repair. Genes (Basel). 12:13902021. View Article : Google Scholar

90 

Morati F and Modesti M: Insights into the control of RAD51 nucleoprotein filament dynamics from single-molecule studies. Curr Opin Genet Dev. 71:182–187. 2021. View Article : Google Scholar : PubMed/NCBI

91 

Wang SSY, Jie YE, Cheng SW, Ling GL and Ming HVY: PARP inhibitors in breast and ovarian cancer. Cancers (Basel). 15:23572023. View Article : Google Scholar : PubMed/NCBI

92 

Konecny GE and Kristeleit RS: PARP inhibitors for BRCA1/2-mutated and sporadic ovarian cancer: Current practice and future directions. Br J Cancer. 115:1157–1173. 2016. View Article : Google Scholar : PubMed/NCBI

93 

Dilmac S and Ozpolat B: Mechanisms of PARP-inhibitor-resistance in BRCA-mutated breast cancer and new therapeutic approaches. Cancers (Basel). 15:36422023. View Article : Google Scholar : PubMed/NCBI

94 

Ghandali M, Huntington K, Srinivasan P, Dizon DS, Graff SL, Carneiro BA and El-Deiry WS: Abstract 1066: PARP inhibitor rucaparib in combination with imipridones ONC201 or ONC212 demonstrates preclinical synergy against BRCA1/2-deficient breast, ovarian and prostate cancer cells. Cancer Res. 83(7_Suppl): S10662023. View Article : Google Scholar

95 

Vidula N, Damodaran S, Bhave M, Rugo H, Shah AN, Blouch E, Ruffle-Deignan NR, Ogbenna O, Flaum LE, Cristofanilli M, et al: Abstract PO4-19-06: Phase II study of a PARP inhibitor, talazoparib, in HER2-metastatic breast cancer with a somatic BRCA1/2 mutation present in cell-free DNA or tumor tissue genotyping. Cancer Res. 84(9_Suppl): PO4-19-062024. View Article : Google Scholar

96 

Vidula N, Blouch E, Basile E, Ruffle-Deignan NR, Horick N, Damodaran S, Aspitia AM, Bhave M, Shah A, Liu MC, et al: Abstract OT2-24-03: Phase II study of a PARP inhibitor in metastatic breast cancer with somatic BRCA1/2 mutations identified by cell-free DNA: Genotyping based clinical trial. Cancer Res. 82(4_Suppl): OT2-24-032024. View Article : Google Scholar

97 

Baldock RA, Pressimone CA, Baird JM, Khodakov A, Luong TT, Grundy MK, Smith CM, Karpenshif Y, Bratton-Palmer DS, Prakash R, et al: RAD51D splice variants and cancer-associated mutations reveal XRCC2 interaction to be critical for homologous recombination. DNA Repair (Amst). 76:99–107. 2019. View Article : Google Scholar : PubMed/NCBI

98 

Castroviejo-Bermejo M, Cruz C, Llop-Guevara A, Gutiérrez-Enríquez S, Ducy M, Ibrahim YH, Gris-Oliver A, Pellegrino B, Bruna A, Guzman M, et al: A RAD51 assay feasible in routine tumor samples calls PARP inhibitor response beyond BRCA mutation. EMBO Mol Med. 10:e91722018. View Article : Google Scholar : PubMed/NCBI

99 

Pelttari LM, Khan S, Vuorela M, Kiiski JI, Vilske S, Nevanlinna V, Ranta S, Schleutker J, Winqvist R, Kallioniemi A, et al: RAD51B in familial breast cancer. PLoS One. 11:e01537882016. View Article : Google Scholar : PubMed/NCBI

100 

Setton J, Selenica P, Mukherjee S, Shah R, Pecorari I, McMillan B, Pei IX, Kemel Y, Ceyhan-Birsoy O, Sheehan M, et al: Germline RAD51B variants confer susceptibility to breast and ovarian cancers deficient in homologous recombination. NPJ Breast Cancer. 7:1352021. View Article : Google Scholar :

101 

Boni J, Idani A, Roca C, Feliubadaló L, Tomiak E, Weber E, Foulkes WD, Orthwein A, El Haffaf Z, Lázaro C and Rivera B: A decade of RAD51C and RAD51D germline variants in cancer. Hum Mutat. 43:285–298. 2021. View Article : Google Scholar : PubMed/NCBI

102 

Kolinjivadi AM, Chong ST, Choudhary R, Sankar H, Chew EL, Yeo C, Chan SH and Ngeow J: Functional analysis of germline RAD51C missense variants highlight the role of RAD51C in replication fork protection. Hum Mol Genet. 32:1401–1409. 2023. View Article : Google Scholar

103 

Yang X, Song H, Leslie G, Engel C, Hahnen E, Auber B, Horváth J, Kast K, Niederacher D, Turnbull C, et al: Ovarian and breast cancer risks associated with pathogenic variants in RAD51C and RAD51D. J Natl Cancer Inst. 112:1242–1250. 2020. View Article : Google Scholar : PubMed/NCBI

104 

Suszyńska M, Ratajska M and Kozlowski P: BRIP1, RAD51C and RAD51D mutations are associated with high susceptibility to ovarian cancer: Mutation prevalence and precise risk estimates based on a pooled analysis of ~30,000 cases. J Ovarian Res. 13:502020. View Article : Google Scholar

105 

Wesoła M and Jeleń M: The risk of breast cancer due to PALB2 gene mutations. Adv Clin Exp Med. 26:339–342. 2017. View Article : Google Scholar

106 

Ruberu TLM, Braun D, Parmigiani G and Biswas S: Meta-analysis of breast cancer risk for individuals with PALB2 pathogenic variants. Genet Epidemiol. 48:448–454. 2024. View Article : Google Scholar : PubMed/NCBI

107 

Sato K, Koyasu M, Nomura S, Sato Y, Kita M, Ashihara Y, Adachi Y, Ohno S, Iwase T, Kitagawa D, et al: Mutation status of RAD 51C, PALB 2 and BRIP 1 in 100 Japanese familial breast cancer cases without BRCA 1 and BRCA 2 mutations. Cancer Sci. 108:2287–2294. 2017. View Article : Google Scholar : PubMed/NCBI

108 

Feng Y, Wang D, Xiong L, Zhen G and Tan J: Predictive value of RAD51 on the survival and drug responsiveness of ovarian cancer. Cancer Cell Int. 21:2492021. View Article : Google Scholar

109 

Alizzi Z, Saravi S, Khalique S, McDonald T, Karteris E and Hall M: Identification of RAD51 foci in cancer-associated circulating cells of patients with high-grade serous ovarian cancer: Association with treatment outcomes. Int J Gynecol Cancer. 33:1427–1433. 2023. View Article : Google Scholar :

110 

Compadre AJ, Van Biljon L, Valentine MC, Llop-Guevara A, Graham E, Fashemi B, Herencia-Ropero A, Kotnik EN, Cooper I, Harrington SP, et al: RAD51 Foci as a biomarker predictive of platinum chemotherapy response in ovarian cancer. Clin Cancer Res. 29:2466–2479. 2023. View Article : Google Scholar : PubMed/NCBI

111 

Goel N, Foxall ME, Scalise CB, Wall JA and Arend RC: Strategies in overcoming homologous recombination proficiency and PARP inhibitor resistance. Mol Cancer Ther. 20:1542–1549. 2021. View Article : Google Scholar

112 

McMullen M, Karakasis K, Madariaga A and Oza AM: Overcoming platinum and PARP-inhibitor resistance in ovarian cancer. Cancers (Basel). 12:16072020. View Article : Google Scholar : PubMed/NCBI

113 

Song H, Dicks E, Ramus SJ, Tyrer JP, Intermaggio MP, Hayward J, Edlund CK, Conti D, Harrington P, Fraser L, et al: Contribution of germline mutations in the RAD51B, RAD51C and RAD51D genes to ovarian cancer in the population. J Clin Oncol. 33:2901–2907. 2015. View Article : Google Scholar :

114 

Nguyen L, WM Martens J, Van Hoeck A and Cuppen E: Pan-cancer landscape of homologous recombination deficiency. Nat Commun. 11:55842020. View Article : Google Scholar :

115 

Nickols NG, Maxwell KN, Lee KM, Hausler R, Anglin-Foote T, Garraway I and Lynch JA: Frequencies of actionable alterations found by somatic tumor sequencing in veterans with metastatic prostate cancer. J Clin Oncol. 40(6_suppl): S1782022. View Article : Google Scholar

116 

Rajput M, Singh R, Singh N and Singh RP: EGFR-mediated RAD51 expression potentiates intrinsic resistance in prostate cancer via EMT and DNA repair pathways. Life Sci. 286:1200312021. View Article : Google Scholar

117 

Maranto C, Udhane V, Hoang DT, Gu L, Alexeev V, Malas K, Cardenas K, Brody JR, Rodeck U, Bergom C, et al: STAT5A/B blockade sensitizes prostate cancer to radiation through inhibition of RAD51 and DNA repair. Clin Cancer Res. 24:1917–1931. 2018. View Article : Google Scholar : PubMed/NCBI

118 

Arce-Gallego S, Llop-Guevara A, Carreira S, Porta N, Fasani R, Bianchini D, Seed G, Rescigno P, Paschalis A, Bertan C, et al: Abstract CT161: A homologous recombination repair (HRR) functional assay to stratify patients with metastatic prostate cancer for PARP inhibitor treatment in the TOPARP-B clinical trial. Cancer Res. 81(Suppl 13): CT1612021. View Article : Google Scholar

119 

Mason JM, Logan HL, Budke B, Wu M, Pawłowski M, Weichselbaum RR, Kozikowski AP, Bishop DK and Connell PP: The RAD51-stimulatory compound RS-1 can exploit the RAD51 overexpression that exists in cancer cells and tumors. Cancer Res. 74:3546–3555. 2014. View Article : Google Scholar : PubMed/NCBI

120 

Zhang Y, Park JY, Zhang F, Olson SH, Orlow I, Li Y, Kurtz RC, Ladanyi M, Chen J, Toland AE, et al: The p. Ser64Leu and p.Pro104Leu missense variants of PALB2 identified in familial pancreatic cancer patients compromise the DNA damage response. Hum Mutat. 42:150–163. 2021. View Article : Google Scholar

121 

Nagathihalli NS and Nagaraju G: RAD51 as a potential biomarker and therapeutic target for pancreatic cancer. Biochim Biophys Acta. 1816:209–218. 2011.

122 

Zhang X, Ma N, Yao W, Li S and Ren Z: RAD51 is a potential marker for prognosis and regulates proliferation in pancreatic cancer. Cancer Cell Int. 19:3562019. View Article : Google Scholar

123 

COSMIC (Catalogue Of Somatic Mutations In Cancer). Sanger Institute. https://cancer.sanger.ac.uk/cosmic/gene/analysis. Note: Access to the specific data referenced requires logging into the COSMIC database. Credentials can be provided upon reasonable request to the corresponding author.

124 

Yang X, Leslie G, Doroszuk A, Schneider S, Allen J, Decker B, Dunning AM, Redman J, Scarth J, Plaskocinska I, et al: Cancer risks associated with germline PALB2 pathogenic variants: an international study of 524 families. J Clin Oncol. 38:674–685. 2020. View Article : Google Scholar

125 

Tischkowitz M, Balmaña J, Foulkes WD, James P, Ngeow J, Schmutzler R, Voian N, Wick MJ, Stewart DR and Pal T; ACMG Professional Practice and Guidelines Committee: Management of individuals with germline variants in PALB2: A clinical practice resource of the American College of Medical Genetics and Genomics (ACMG). Genet Med. 23:1416–1423. 2021. View Article : Google Scholar : PubMed/NCBI

126 

Kwong A, Ho CYS, Au CH, Tey SK and Ma ESK: Germline RAD51C and RAD51D mutations in high-risk Chinese breast and/ or ovarian cancer patients and families. J Pers Med. 14:8662024. View Article : Google Scholar

127 

Lee A, Mavaddat N, Wilcox AN, Cunningham AP, Carver T, Hartley S, Babb de Villiers C, Izquierdo A, Simard J, Schmidt MK, et al: BOADICEA: A comprehensive breast cancer risk prediction model incorporating genetic and nongenetic risk factors. Genet Med. 21:1708–1718. 2019. View Article : Google Scholar : PubMed/NCBI

128 

Natiaonal Cancer Institute: Genetic: Testing for Inherited Cancer Risk. https://www.cancer.gov/about-cancer/causes-prevention/genetics/genetic-testing-fact-sheet. Accessed April 18, 2024

129 

Faucett WA, Peay H and Coghlin CR II: Genetic testing: Consent and result disclosure for the primary care provider. Med Clin North Am. 103:967–976. 2019. View Article : Google Scholar :

130 

Al-Shamsi HO, Alwbari A, Azribi F, Calaud F, Thuruthel S, Tirmazy SHH, Kullab S, Ostomane S and Abulkhair O: BRCA testing and management of BRCA-mutated early-stage breast cancer: A comprehensive statement by expert group from GCC region. Front Oncol. 14:13589822024. View Article : Google Scholar : PubMed/NCBI

131 

Natiaonal Cancer Institute: Surgery to Reduce the Risk of Breast Cancer. https://www.cancer.gov/types/breast/risk-reducing-surgery-fact-sheet#:~:text=Risk%2Dreducing%20salpingo%2Doophorectomy%20greatly,gene%20(23%E2%80%9325). Accessed June 26, 2024

132 

Mai PL, Miller A, Gail MH, Skates S, Lu K, Sherman ME, Ioffe OB, Rodriguez G, Cohn DE, Boggess J, et al: Risk-reducing salpingo-oophorectomy and breast cancer risk reduction in the Gynecologic Oncology Group Protocol-0199 (GOG-0199). JNCI Cancer Spectr. 4:pkz0752019. View Article : Google Scholar

133 

Wang Y, Song Z, Zhang S, Wang X and Li P: Risk-reducing salpingo-oophorectomy and breast cancer risk in BRCA1 or BRCA2 mutation carriers: A systematic review and meta-analysis. Eur J Surg Oncol. 48:1209–1216. 2022. View Article : Google Scholar

134 

Oceguera-Basurto P, Topete A, Oceguera-Villanueva A, Rivas-Carrillo J, Paz-Davalos M, Quintero-Ramos A and Daneri-Navarro A: Selective estrogen receptor modulators in the prevention of breast cancer in premenopausal women: A review. Transl Cancer Res. 9:4444–4456. 2020. View Article : Google Scholar : PubMed/NCBI

135 

Peters A and Tadi P: Aromatase inhibitors. StatPearls [Internet]. StatPearls Publishing; Treasure Island, FL: 2023

136 

American Cancer Society (ACS): Aromatase Inhibitors for Lowering Breast Cancer Risk. ACS; Atlanta, GA: 2021, https://www.cancer.org/cancer/types/breast-cancer/risk-and-preven-tion/aromatase-inhibitors-for-lowering-breast-cancer-risk.html.

137 

Lord CJ and Ashworth A: PARP inhibitors: Synthetic lethality in the clinic. Science. 355:1152–1158. 2017. View Article : Google Scholar

138 

Galland L, Ballot E, Mananet H, Boidot R, Lecuelle J, Albuisson J, Arnould L, Desmoulins I, Mayeur D, Kaderbhai C, et al: Efficacy of platinum-based chemotherapy in metastatic breast cancer and HRD biomarkers: Utility of exome sequencing. NPJ Breast Cancer. 8:282022. View Article : Google Scholar : PubMed/NCBI

139 

Qiu Z, Oleinick NL and Zhang J: ATR/CHK1 inhibitors and cancer therapy. Radiother Oncol. 126:450–464. 2018. View Article : Google Scholar

140 

Biegała Ł, Gajek A, Szymczak-Pajor I, Marczak A, Śliwińska A and Rogalska A: Targeted inhibition of the ATR/CHK1 pathway overcomes resistance to olaparib and dysregulates DNA damage response protein expression in BRCA2 MUT ovarian cancer cells. Sci Rep. 13:226592023. View Article : Google Scholar

141 

Ha DH, Min A, Kim S, Jang H, Kim SH, Kim HJ, Ryu HS, Ku JL, Lee KH and Im SA: Antitumor effect of a WEE1 inhibitor and potentiation of olaparib sensitivity by DNA damage response modulation in triple-negative breast cancer. Sci Rep. 10:99302020. View Article : Google Scholar :

142 

Jiang X, Bai H, Li X, Li W and Zhang Z: Current status and future prospects of PARP inhibitor clinical trials in ovarian cancer. Cancer Manag Res. 11:4371–4390. 2019. View Article : Google Scholar : PubMed/NCBI

143 

Liposits G, Loh KP, Soto-Perez-de-Celis E, Dumas L, Battisti NML, Kadambi S, Baldini C, Banerjee S and Lichtman SM: PARP inhibitors in older patients with ovarian and breast cancer: Young International Society of Geriatric Oncology review paper. J Geriatr Oncol. 10:337–345. 2019. View Article : Google Scholar

144 

Boussios S, Moschetta M, Karihtala P, Samartzis EP, Sheriff M, Pappas-Gogos G, Ozturk MA, Uccello M, Karathanasi A, Tringos M, et al: Development of new poly(ADP-ribose) polymerase (PARP) inhibitors in ovarian cancer: Quo Vadis? Ann Transl Med. 8:17062020. View Article : Google Scholar

145 

Boussios S, Karihtala P, Moschetta M, Karathanasi A, Rassy E, Sadauskaite A and Pavlidis N: Combined strategies with poly (ADP-Ribose) polymerase (PARP) inhibitors for the treatment of ovarian cancer: A literature review. Diagnostics (Basel). 9:872019. View Article : Google Scholar : PubMed/NCBI

146 

Plummer R: Poly(ADP-ribose) polymerase inhibition: A new direction for BRCA and triple-negative breast cancer? Breast Cancer Res. 13:2182011. View Article : Google Scholar : PubMed/NCBI

147 

Boussios S, Abson C, Moschetta M, Rassy E, Karathanasi A, Bhat T, Ghumman F, Sheriff M and Pavlidis N: Poly (ADP-Ribose) polymerase inhibitors: Talazoparib in ovarian cancer and beyond. Drugs R D. 20:55–73. 2020. View Article : Google Scholar : PubMed/NCBI

148 

O'Sullivan Coyne G, Chen A and Kummar S: Delivering on the promise: Poly ADP ribose polymerase inhibition as targeted anticancer therapy. Curr Opin Oncol. 27:475–481. 2015. View Article : Google Scholar

149 

Mittica G, Ghisoni E, Giannone G, Genta S, Aglietta M, Sapino A and Valabrega G: PARP inhibitors in ovarian cancer. Recent Pat Anticancer Drug Discov. 13:392–410. 2018. View Article : Google Scholar : PubMed/NCBI

150 

Zimmer AS, Gillard M, Lipkowitz S and Lee JM: Update on PARP inhibitors in breast cancer. Curr Treat Options Oncol. 19:212018. View Article : Google Scholar

151 

Alva AS, Mangat PK, Garrett-Mayer E, Halabi S, Hansra D, Calfa CJ, Khalil MF, Ahn ER, Cannon TL, Crilley P, et al: Pembrolizumab in patients with metastatic breast cancer with high tumor mutational burden: Results from the targeted agent and profiling utilization registry (TAPUR) study. J Clin Oncol. 39:2443–2451. 2021. View Article : Google Scholar : PubMed/NCBI

152 

Cortesi L, Rugo HS and Jackisch C: An Overview of PARP inhibitors for the treatment of breast cancer. Target Oncol. 16:255–282. 2021. View Article : Google Scholar

153 

Jenner ZB, Sood AK and Coleman RL: Evaluation of rucaparib and companion diagnostics in the PARP inhibitor landscape for recurrent ovarian cancer therapy. Future Oncol. 12:1439–1456. 2016. View Article : Google Scholar

154 

Slootbeek PHJ, Overbeek JK, Ligtenberg MJL, Van Erp NP and Mehra N: PARPing up the right tree; an overview of PARP inhibitors for metastatic castration-resistant prostate cancer. Cancer Lett. 577:2163672023. View Article : Google Scholar : PubMed/NCBI

155 

Cruz C, Castroviejo-Bermejo M, Gutiérrez-Enríquez S, Llop-Guevara A, Ibrahim YH, Gris-Oliver A, Bonache S, Morancho B, Bruna A, Rueda OM, et al: RAD51 foci as a functional biomarker of homologous recombination repair and PARP inhibitor resistance in germline BRCA-mutated breast cancer. Ann Oncol. 29:1203–1210. 2018. View Article : Google Scholar

156 

Orhan E, Velázquez C, Tabet I, Sardet C and Theillet C: Regulation of RAD51 at the transcriptional and functional levels: What prospects for cancer therapy? Cancers (Basel). 13:29302021. View Article : Google Scholar : PubMed/NCBI

157 

Zhao L, Si CS, Yu Y, Lu JW and Zhuang Y: Depletion of DNA damage binding protein 2 sensitizes triple-negative breast cancer cells to poly ADP-ribose polymerase inhibition by destabilizing RAD51. Cancer Sci. 110:3543–3552. 2019. View Article : Google Scholar : PubMed/NCBI

158 

Jia Y, Song Y, Dong G, Hao C, Zhao W, Li S and Tong Z: Aberrant regulation of RAD51 promotes resistance of neoadjuvant endocrine therapy in ER-positive breast cancer. Sci Rep. 9:129392019. View Article : Google Scholar : PubMed/NCBI

159 

Goričar K, Dugar F, Dolžan V and Marinko T: NBN, RAD51 and XRCC3 polymorphisms as potential predictive biomarkers of adjuvant radiotherapy toxicity in early HER2-positive breast cancer. Cancers (Basel). 14:43652022. View Article : Google Scholar

160 

Yu J and Wang CG: Relationship between polymorphisms in homologous recombination repair genes RAD51 G172T, XRCC2 & XRCC3 and risk of breast cancer: A meta-analysis. Front Oncol. 13:10473362023. View Article : Google Scholar

161 

Day M, Lapierre J, O'Shea T and Mills K: Abstract C14: A novel RAD51 inhibitor, CYT-0851, shows anticancer activity in preclinical models of pancreatic cancer. Cancer Res. 79(24_Suppl): C142019. View Article : Google Scholar

162 

Tsai YF, Chan LP, Chen YK, Su CW, Hsu CW, Wang YY and Yuan SF: RAD51 is a poor prognostic marker and a potential therapeutic target for oral squamous cell carcinoma. Cancer Cell Int. 23:2312023. View Article : Google Scholar : PubMed/NCBI

163 

Korsholm LM, Kjeldsen M, Perino L, Mariani L, Nyvang GB, Kristensen E, Bagger FO, Mirza MR and Rossing M: Combining homologous recombination-deficient testing and functional RAD51 analysis enhances the prediction of poly(ADP-Ribose) polymerase inhibitor sensitivity. JCO Precis Oncol. 8:e23004832024. View Article : Google Scholar :

164 

QI AGEN: Browse the manual. Calculate HRD Score (beta). https://resources.qiagenbioinformatics.com/manuals/biomedicalgenomicsanalysis/current/index.php?manual=Calculate_HRD_Score_beta.html.

165 

van Wijk LM, Nilas AB, Vrieling H and Vreeswijk MPG: RAD51 as a functional biomarker for homologous recombination deficiency in cancer: A promising addition to the HRD toolbox? Expert Rev Mol Diagn. 22:185–199. 2022. View Article : Google Scholar

166 

Vogel A, Haupts A, Kloth M, Roth W and Hartmann N: A novel targeted NGS panel identifies numerous homologous recombination deficiency (HRD)-associated gene mutations in addition to known BRCA mutations. Diagn Pathol. 19:92024. View Article : Google Scholar : PubMed/NCBI

167 

Witz A, Dardare J, Betz M, Michel C, Husson M, Gilson P, Merlin JL and Harlé A: Homologous recombination deficiency (HRD) testing landscape: Clinical applications and technical validation for routine diagnostics. Biomark Res. 13:312025. View Article : Google Scholar : PubMed/NCBI

168 

Xu Y, Chen YA, Wu Y, Saverimuthu A, Jadhav A, Bhuiyan R, Sandler J, Yio J and Kumar V: The prognostic and predictive value of homologous recombination deficiency status in patients with advanced stage epithelial ovarian carcinoma after first-line platinum-based chemotherapy. Front Oncol. 14:13724822024. View Article : Google Scholar :

169 

Pellegrino B, Herencia-Ropero A, Llop-Guevara A, Pedretti F, Moles-Fernández A, Viaplana C, Villacampa G, Guzmán M, Rodríguez O, Grueso J, et al: Preclinical in vivo validation of the RAD51 test for identification of homologous recombination-deficient tumors and patient stratification. Cancer Res. 82:16462022. View Article : Google Scholar

170 

Guffanti F, Mengoli I and Damia G: Current HRD assays in ovarian cancer: Differences, pitfalls, limitations and novel approaches. Front Oncol. 14:14053612024. View Article : Google Scholar

171 

Zhou J, Wang H, Fu F, Li Z, Feng Q, Wu W, Liu Y, Wang C and Chen Y: Spectrum of PALB2 germline mutations and characteristics of PALB2-related breast cancer: Screening of 16,501 unselected patients with breast cancer and 5890 controls by next-generation sequencing. Cancer. 126:3202–3208. 2020. View Article : Google Scholar

172 

Woods NT, Baskin R, Golubeva V, Jhuraney A, De-Gregoriis G, Vaclova T, Goldgar DE, Couch FJ, Carvalho MA, Iversen ES and Monteiro AN: Functional assays provide a robust tool for the clinical annotation of genetic variants of uncertain significance. NPJ Genom Med. 1:160012016. View Article : Google Scholar : PubMed/NCBI

173 

Rein HL and Bernstein KA: Finding significance: New perspectives in variant classification of the RAD51 regulators, BRCA2 and beyond. DNA Repair (Amst). 130:1035632023. View Article : Google Scholar : PubMed/NCBI

174 

Zielli T, Labidi-Galy I, Del Grande M, Sessa C and Colombo I: The clinical challenges of homologous recombination proficiency in ovarian cancer: From intrinsic resistance to new treatment opportunities. Cancer Drug Resist. 6:499–516. 2023. View Article : Google Scholar :

175 

Li A, Geyer FC, Blecua P, Lee JY, Selenica P, Brown DN, Pareja F, Lee SSK, Kumar R, Rivera B, et al: Homologous recombination DNA repair defects in PALB2-associated breast cancers. NPJ Breast Cancer. 5:232019. View Article : Google Scholar :

Related Articles

Journal Cover

August-2025
Volume 67 Issue 2

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kausar MA, Alshammari KF, Alenazi F, Anwar S, Khalifa AM, Ginawi T, Asiri A, Najm MZ, Rabbani SA, El‑Tanani M, El‑Tanani M, et al: <em>RAD51</em> and <em>PALB2</em> in precision oncology: Clinical implications for HRD associated breast and ovarian cancers (Review). Int J Oncol 67: 65, 2025.
APA
Kausar, M.A., Alshammari, K.F., Alenazi, F., Anwar, S., Khalifa, A.M., Ginawi, T. ... Gantayat, S. (2025). <em>RAD51</em> and <em>PALB2</em> in precision oncology: Clinical implications for HRD associated breast and ovarian cancers (Review). International Journal of Oncology, 67, 65. https://doi.org/10.3892/ijo.2025.5771
MLA
Kausar, M. A., Alshammari, K. F., Alenazi, F., Anwar, S., Khalifa, A. M., Ginawi, T., Asiri, A., Najm, M. Z., Rabbani, S. A., El‑Tanani, M., Gantayat, S."<em>RAD51</em> and <em>PALB2</em> in precision oncology: Clinical implications for HRD associated breast and ovarian cancers (Review)". International Journal of Oncology 67.2 (2025): 65.
Chicago
Kausar, M. A., Alshammari, K. F., Alenazi, F., Anwar, S., Khalifa, A. M., Ginawi, T., Asiri, A., Najm, M. Z., Rabbani, S. A., El‑Tanani, M., Gantayat, S."<em>RAD51</em> and <em>PALB2</em> in precision oncology: Clinical implications for HRD associated breast and ovarian cancers (Review)". International Journal of Oncology 67, no. 2 (2025): 65. https://doi.org/10.3892/ijo.2025.5771