metricas
covid
Buscar en
Revista de Senología y Patología Mamaria - Journal of Senology and Breast Disease
Toda la web
Inicio Revista de Senología y Patología Mamaria - Journal of Senology and Breast Dise... Understanding the prospective of gene therapy for the treatment of breast cancer
Información de la revista
Vol. 38. Núm. 4. (En progreso)
(octubre - diciembre 2025)
Visitas
2134
Vol. 38. Núm. 4. (En progreso)
(octubre - diciembre 2025)
Review
Acceso a texto completo
Understanding the prospective of gene therapy for the treatment of breast cancer
Comprensión de las perspectivas de la terapia génica para el tratamiento del cáncer de mama
Visitas
2134
Ritikaa, Sonam Rania, Rishabha Malviyab,
Autor para correspondencia
, Shivam Rajputb, Sathvik Belagodu Sridharc, Dinesh Kaushika
a Hindu College of Pharmacy, Gohana Road, Sonipat, Haryana, India
b Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
c RAK College of Pharmacy, RAK Medical & Health Sciences University, Ras Al Khaimah, United Arab Emirates
Este artículo ha recibido
Información del artículo
Resumen
Texto completo
Bibliografía
Descargar PDF
Estadísticas
Figuras (6)
Mostrar másMostrar menos
Abstract

Breast cancer (BC) is the most common type of cancer in women and the second most common cause of death from cancer in women around the world. Local therapies, Systematic therapies and advanced systematic therapies are included in current treatment strategies. Gene therapy has become a hopeful way to treat or stop disease by changing bad genes genetically. This innovative strategy aims to replace defective genes, enhance immune defenses, or silence harmful genes. Recent advances in cancer-specific promoter systems make targeted gene treatments possible. These include using suicide genes, cancer suppressor genes, gene silencing technologies, and gene-editing techniques. Among these, CRISPR and CAR-T therapy have revolutionized genetic editing in cancer research, offering high specificity, precision, cost-effectiveness, and efficiency. This review highlights recent advancements in gene therapy targeting cancer-specific promoters, emphasizing its potential to transform BC treatment.

Keywords:
Breast cancer
Tumor
Gene therapy
Gene editing
CRISPR
CAR-T cells
Resumen

El cáncer de mama (CM) es la segunda causa de muerte relacionada con el cáncer en mujeres en todo el mundo, así como la neoplasia maligna más comúnmente diagnosticada. Para abordarlo, se han desarrollado una variedad de opciones de tratamiento, que incluyen cirugía, radioterapia, terapia hormonal, quimioterapia y terapias biológicas. La terapia génica ha despertado interés como una forma novedosa de tratar muchos tipos de cáncer mediante la modificación de genes defectuosos. La terapia génica es el proceso de alterar genéticamente los genes de una célula para tratar o prevenir enfermedades. Busca tratar enfermedades, aumentar la capacidad del sistema inmunológico para defenderse de las enfermedades o cambiar genes dañados por otros funcionales. Esta tecnología se utiliza para alterar genéticamente las células del paciente para mejorar o tratar su condición. Se han creado varios sistemas promotores específicos del cáncer para atacar las células cancerosas. Busca proporcionar información actualizada sobre la terapia génica dirigida a promotores específicos del cáncer. Esto fomenta ciertas terapias, como genes supresores del cáncer, genes suicidas, silenciamiento de genes y tratamientos de edición de genes. La tecnología CRISPR, un descubrimiento clave en la edición genética, ahora se utiliza ampliamente en terapia génica y en la investigación del cáncer humano. Es ampliamente considerado por su bajo riesgo, sus cualidades de ahorro de tiempo, su rentabilidad, su especificidad y su precisión.

Palabras clave:
Cáncer de mama
Tumor
Terapia génica
Terapia avanzada
Edición de genes
CRISPR
Células CAR-T
Texto completo
Introduction

Breast cancer (BC) is the most common cancer in women and the second most common cause of death from cancer in the world.1 There are glandular and stromal cells in the breast. A glandular tissue has lobules that make milk and tubes that carry milk to the nipple.2 The stromal tissue comprises fatty and fibrous connective tissues, and the lymphatic tissue aids in removing cellular fluids and waste products through the immune system.3,4 BC can develop in various parts of the breast, with ductal carcinomas being the most common type, originating in the cells lining the milk ducts. Lobular carcinomas, arising from the lobules, are less common but still significant.5,6 While most changes in the breast are benign, malignant transformations, though rare, can occur in breast cells, with ductal and lobular carcinomas being the primary forms of BC.7

The causes of BC are multifactorial, involving both intrinsic and extrinsic factors. Lifestyle, environmental, psychological, and social factors contribute significantly to its prevalence. About 20–30% of BC cases are caused by risk factors that can be changed. The other 5–10% are caused by genetic changes or a family history of the disease.8,9 With 2.08 million new cases and 626,000 deaths in 2018, BC was responsible for 6.6% of all cancer-related deaths around the world.10–12 Several lifestyle factors increase BC risk, including obesity, physical inactivity, and alcohol consumption. Hormonal and reproductive factors such as early menarche, delayed childbirth, fewer pregnancies, lack of breastfeeding, and the use of hormonal therapies or oral contraceptives also contribute to the risk.13–19 Genetic factors are very important. Changes in high-risk genes like BRCA1, BRCA2, PALB2, ATM, CHEK2, and TP53 make people much more likely to get BC.20–22

Current treatment modules for BC

Various treatment modules for breast cancer are summarized in Fig. 1.

Figure 1.

Current treatment modules for breast cancer.

(0.32MB).
Local therapies

Local therapy for BC is an essential component of the therapeutic strategy, designed to specifically target cancer cells in the breast and adjacent tissues while reducing effects on the remainder of the body.23,24 The primary modalities of local therapy include surgical intervention and radiation therapy, frequently employed in conjunction with systemic treatments such as chemotherapy, hormone therapy, or targeted therapy, contingent upon the particularities of the malignancy. Surgery is generally the primary local intervention for BC. The selection of a surgical technique is contingent upon aspects including tumor size and location, alongside the patient's preferences and overall health status. The two principal categories of surgical procedures are lumpectomy and mastectomy.25,26 A lumpectomy entails excising the tumor together with a narrow margin of adjacent healthy tissue. This operation is commonly known as breast-conserving surgery, as it preserves the majority of the breast tissue. After a lumpectomy, the majority of patients will have radiation therapy to eradicate any residual cancer cells that may be undetectable yet provide a risk of recurrence. Alongside these surgical alternatives, radiation therapy is essential in localized treatment.27–29 Radiation employs high-energy photons, including X-rays, to precisely target and eradicate cancer cells. Post-surgery, radiation therapy is frequently employed to diminish the probability of cancer recurrence, particularly in instances involving lumpectomy or when there is a danger of residual cancer cells in the breast or adjacent lymph nodes.30,31 Radiation may be advised for patients with bigger tumors or those whose cancer has metastasized to the axillary lymph nodes. The process entails directing controlled radiation treatments at the affected area over several weeks, generally five days per week, while the specific treatment plan is contingent upon individual conditions.32,33

Systematic therapies

Systemic therapy for BC is a treatment modality that addresses cancer cells throughout the body, rather than focusing solely on the breast tumor. This therapy is particularly crucial for addressing malignancies that may have metastasized beyond the breast, affecting adjacent lymph nodes or distant organs. The primary forms of systemic therapy encompass chemotherapy, hormone therapy, targeted therapy, and immunotherapy. Chemotherapy employs potent pharmaceuticals to eradicate rapidly proliferating neoplastic cells. It may be provided before surgery (neoadjuvant therapy) to diminish tumor size, after surgery (adjuvant therapy) to mitigate recurrence risk, or in instances of metastatic BC to manage the disease.34–38 Hormone therapy is employed for hormone receptor-positive breast tumors, in which tumor cells depend on hormones such as estrogen or progesterone for proliferation. Medications such as tamoxifen or aromatase inhibitors inhibit or diminish these hormones, aiding in the prevention of cancer recurrence.39,40 Targeted therapy concentrates on particular attributes of cancer cells, exemplified by HER2-positive tumors, where agents such as trastuzumab (Herceptin) specifically inhibit the HER2 protein on cancer cells to impede their proliferation. Immunotherapy enhances the body's immune system to more effectively identify and combat cancer cells and is frequently employed for specific aggressive types of BC, such as triple-negative BC.31,41

Advanced systemic therapies

Advanced systemic therapies, such as Antibody-Drug Conjugates (ADCs), immune checkpoint inhibitors and Selective estrogen receptor degraders (SERDs) or emerging technologies like PROTACs exemplify innovative strategies in the management of advanced BC, providing renewed optimism for patients with metastatic or treatment-resistant conditions.

Antibody-drug conjugates (ADCs) therapy

Antibody-drug conjugates (ADCs), exemplified as trastuzumab deruxtecan (Enhertu), integrate the specificity of monoclonal antibodies with the efficacy of chemotherapeutic agents. Trastuzumab deruxtecan is an antibody that specifically targets the HER2 protein, which is overexpressed in certain breast tumors.31,42 Upon binding to the HER2 receptors on cancer cells, the antibody administers a chemotherapeutic agent directly to the tumor. This facilitates a more focused assault on cancer cells while reducing harm to adjacent healthy tissues, rendering it particularly efficacious in HER2-positive BC that has developed resistance to alternative therapies. ADCs provide a more accurate and potent method for cancer treatment, enhancing response rates in individuals with metastatic or previously resistant BC.43–46

Immune checkpoint inhibitors

Immune checkpoint inhibitors, including pembrolizumab (Keytruda) and atezolizumab (Tecentriq), represent a groundbreaking class of pharmaceuticals that function by augmenting the body's immune system to more effectively identify and target cancer cells. Cancer cells frequently avoid immune system identification via immunological checkpoints, such as PD-L1, which function as inhibitors of immune response.47,48 Immune checkpoint inhibitors obstruct these checkpoints, enhancing the immune system's capacity to identify and eliminate cancer cells. This strategy has demonstrated considerable potential, especially in triple-negative BC (TNBC), which is frequently more aggressive and devoid of targeted medicines such as hormone receptor or HER2-based therapy. These inhibitors enhance the immune system's capacity to combat cancer, resulting in better survival and response rates, particularly when used in conjunction with other therapies.49

Selective Estrogen Receptor Degraders (SERDs) and PROTACs therapies

Selective Estrogen Receptor Degraders (SERDs) and innovative technologies such as PROTACs (Proteolysis-Targeting Chimeras) exemplify advanced methodologies in the management of BC, especially for estrogen receptor-positive (ER+) variants. SERDs, including fulvestrant, function by attaching to the estrogen receptor and facilitating its degradation, so obstructing estrogen signaling that stimulates tumor proliferation.50 SERDs are particularly advantageous in instances where cancer cells have acquired resistance to conventional hormone treatments. Conversely, PROTACs are a novel category of small compounds that utilize the body's intrinsic protein degradation mechanisms to selectively target and eradicate certain proteins, including those implicated in the advancement of BC. PROTACs function by enlisting the cell's ubiquitin-proteasome system to eliminate cancer-related proteins, including mutant variants of the estrogen receptor and other critical molecules implicated in tumor viability.51,52

Advanced gene therapy for BC

Gene therapy involves altering a cell's DNA to treat or prevent diseases.53 Genes, composed of DNA, control many body functions, from growth to vital processes.54 Diseases often arise from faulty genes, and gene therapy aims to replace defective genes with functional ones, address illnesses, or boost the immune system.55,56 This innovative technology modifies a patient's cells to improve or treat their condition.57,58 Introducing therapeutic gene material with regulatory components into the nucleus, either restores the normal expression of a defective gene or recovers lost function due to mutations.59,60 However, implementing this approach requires careful biosafety governance. Assessing and mitigating risks to human and environmental health is crucial, underscoring the importance of stringent oversight in gene therapy applications.61,62

As a result, both viral and nonviral vectors are being used in experimental and clinical settings to change genes and treat diseases.63 By 2019, more than 2600 gene therapy clinical trials had been accepted or concluded. In the US, four gene therapy products have been approved by the FDA.64,65 Of the global 232 approved gene and cell therapy trials, approximately 70% used viral vectors.66 More information about gene therapy for BC will be provided in the next section.

Method of gene therapy

There are two methods for introducing genetic material into cells (Fig. 2). Ex-vivo gene therapy involves taking cells from a patient (autologous) or a volunteer (allogeneic), changing their genes in a lab using a vector, and then putting the changed cells back into the patient after activation or expansion.67,68 In-vivo gene therapy, on the other hand, puts a vector straight into the body of the patient, so cells do not have to be taken out. The advantages and downsides of each method are different.

Figure 2.

Ex vivo and in vivo gene transfer mechanism.

(0.24MB).

Ex-vivo gene transfer allows for precise targeting of specific collected cells, ensuring they possess the desired characteristics and offering multiple vector options for enhancement before transplantation. While this method requires advanced laboratory facilities, extensive research, and modern equipment, it provides greater control over the process. In contrast, in vivo, gene modification requires less lab work and is the preferred method when cells cannot be extracted or maintained outside the body. It is ideal for accessible tissues, though nonspecific interactions between vectors and cells, as well as the potential for immune reactions, limit its use.69–71

Mechanism of action of gene therapy for BCSuicide gene

Suicide gene therapy is designed to induce tumor cell death by introducing specific genes that trigger cytotoxic effects, either directly or indirectly (Fig. 3). In direct suicide gene therapy, genes encoding proteins that cause tumor cell death are introduced. Among these is the diphtheria toxin, which is well known. A gene damages the cell membrane, making tumor cells less likely to survive.72–74 Indirect suicide gene therapy, on the other hand, involves adding genes that make enzymes like Herpes Simplex Virus Thymidine Kinase (HSV-tk), which turn on prodrugs like 5-fluorocytosine (5-FC). This change creates 5-fluorouracil (5-FU), a chemotherapy drug that can help fight cancers of the breast, prostate, stomach, and hepatocellular carcinoma (HCC).75,76 Another method involves synthetic molecules like B/B Homodimer (AP20187) and a modified human Caspase 9 (iCas9) gene, which, when delivered to cancer cells or mesenchymal stromal cells (MSCs), trigger apoptosis by inducing dimerization.77–79 Additionally, therapies targeting specific promoters like the H19 RNA gene can regulate cancer cell proliferation. It has been shown that blocking H19 can cause tumors to shrink and cells to die.80,81 Specifically targeting the hTERT gene, which helps tumor cells live forever, has also shown promise in encouraging tumor regression.82 Therapies such as OBP-301 (Telomelysin) have shown promise in preclinical tests.83

Figure 3.

Schematic illustration of suicide gene therapy mechanism.

(0.19MB).
Gene silencing

Transferring particular small interfering RNA (siRNA) into cells allows targeted gene silencing (Fig. 4). The siRNA forms a duplex with the RNA-induced silencing complex (RISC), which causes messenger RNA (mRNA) breakdown and disturbs protein synthesis, therefore silencing the intended gene. Targeting any gene, siRNA is a potential treatment method for many diseases including cancer, liver cirrhosis, hepatitis B, hypercholesterolemia, and human papillomavirus. Unlike other methods, siRNA does not bind to chromosomal DNA, reducing the risk of gene alterations or mutations. Its high specificity helps prevent drug resistance with minimal side effects. siRNA has demonstrated the ability to suppress cancer-related genes, shrinking tumors without eliminating faulty genes. However, systemic delivery remains challenging due to rapid clearance. To deal with this, delivery systems like CALAA-01 for melanoma and ALN-VSPOI for liver cancer are currently going through clinical studies. However, the approach faces challenges because it is toxic and does not perform effectively at transfection.84–87

Figure 4.

Schematic illustration of gene silencing mechanism.

(0.19MB).
Gene editing techniques for cancer

The advancement of gene editing tools has revolutionized the ability to make precise changes to specific gene sequences in eukaryotic cells, with broad applications in research, clinical settings, and biotechnology (Fig. 5).88 ZFNs, TALENs, and CRISPR/Cas are examples of customizable nucleases that have sped up the move from lab studies to clinical use. ZFNs target DNA sequences by putting together a Cys2-His2 protein and the Fok1 restriction endonuclease domain.89–91 Similarly, TALENs use Fok1's catalytic domain and a DNA-binding domain for targeted cleavage.92 These methods have been successful in inhibiting the proliferation of acute lymphoblastic leukemia and cervical cancer cells,93–95 and addressing treatment resistance in BC cells.95 The CRISPR/Cas system, which is a defense mechanism for bacteria, has made gene editing even better by using guide RNA (gRNA) to target specific DNA sequences and Cas proteins for cleavage. This system has a lot of promise for improving immunity, fighting cancer, and fixing mutations.96,97 CRISPR has shown effectiveness in reducing resistance, tumor size, and metastasis in cancers like pancreatic, prostate, colon, and BC.98–103

Figure 5.

Schematic illustration of gene editing technique mechanism.

(0.27MB).
Directing gene therapy in cancer

Gene therapy has made significant strides in recent years; however, one of the biggest problems still is getting therapeutic nucleic acids (TNAs) to the right cells to make them function.104 Developing a carrier that can specifically target and deliver TNAs to cancer cells, while controlling their expression within those cells, is crucial for ensuring precise and effective treatment. Such a carrier must be specifically designed to transport genetic material efficiently.105,106

Regulatory sequences are another important part of gene therapy. They control gene expression in target cells, even if the recombinant DNA cannot get into healthy cells.107 The success of cancer gene therapy often hinges on these regulatory elements. Promoters, which are cis-acting sequences that control the initiation of mRNA transcription, are critical in ensuring that therapeutic genes are expressed only in the desired tissues.108 Located before the coding region of a gene, promoters bind transcription factors and regulate gene activity.109

Promoters are an active area of biotechnology research due to their ability to enhance transcription and provide additional control over gene expression. They can target specific organs or tissues, helping to control gene production and minimize unwanted side effects. Ideally, a promoter must be both strong enough to ensure robust gene expression and specific enough to limit expression to the desired tissue, ensuring safety and efficacy. Three main categories of promoters tissue-specific, cancer-specific, and tumor-specific can be utilized to precisely target cancer cells at the transcriptional level.110,111

Role of CRISPR/Cas9

CRISPR/Cas9 technology has changed the way researchers study BC by allowing exact gene modifications. These changes have helped us learn a lot more about how tumors grow and cells multiply (Fig. 6). CRISPR, which stands for “Clustered Regularly Interspaced Short Palindromic Repeats” was first found in the IAP gene of Escherichia coli by Yoshizumi Ishino and his coworkers in 1987. Bacteria use it to protect themselves against viruses.112 This finding laid the groundwork for gene-editing applications in molecular biology. In the early 2000s, Dr. Jennifer Doudna and her team adapted the CRISPR system for DNA editing, leading to the development of the CRISPR/Cas9 technology.113 By 2013, the first successful human gene editing was achieved using CRISPR/Cas9,114 marking a pivotal moment in biotechnology. These days, CRISPR/Cas9 is one of the most useful tools for both science and medicine.

Figure 6.

Mechanism of CRISPR/Cas9 gene.

(0.3MB).

The CRISPR/Cas9 system is made up of three main parts: a noncoding RNA (crRNA) that targets the DNA sequence; a tracrRNA that, when combined with crRNA, creates a single-guide RNA (sgRNA); and a Cas protein that cuts the target DNA.115 The CRISPR mechanism allows prokaryotes to acquire foreign DNA fragments from viruses, which are integrated into the host genome as memory sequences known as spacers. These spacers enable bacteria to recognize and disable foreign DNA during subsequent infections.116 The insertion of these foreign DNA sequences into the genome is done by Cas1 and Cas2 proteins, with Cas1 having the appropriate enzyme activity.117–119 Once spacers are added, the CRISPR locus is turned into a long RNA molecule that has both spacer and repeat sequences. At the 3′ end of this pre-crRNA, the tracrRNA binds. This makes a double-stranded RNA that RNase III cuts. This process makes a fully developed gRNA complex that can direct the Cas protein to its target.120–123 The gRNA complex sets off the Cas9 protein, which then looks for foreign DNA sequences that match and cut the DNA at the target spot. This process is what makes CRISPR-mediated defense in bacteria work.124 CRISPR/Cas9 is a very important tool in genetic research because it is easy to use and can do many different things. It is especially useful for learning how diseases like BC are caused genetically. The need for a PAM sequence for targeting is a drawback of CRISPR/Cas9, despite its great efficiency.125 Researchers have come up with Cas9 variants like spCas9 to tackle this issue. These variants can recognize a wider range of PAM sequences, making the mechanism more versatile.126,127 Additionally, modified versions of Cas9, known as deactivated Cas9 (dCas9), are employed in applications such as transcriptional suppression or activation via CRISPR interference (CRISPRi) and CRISPR activation (CRISPRa).128,129 These innovations allow for more precise gene regulation, providing powerful tools for cancer research and therapies, especially in correcting mutations and offering potential treatment options for BC and other diseases.

The conflicting clinical findings suggest that CRISPR technology may serve as an efficient way to validate medication specificity in preclinical research prior to clinical testing. In comparison to other gene modification techniques, such as antagonists or RNA interference (RNAi), CRISPR-mediated gene manipulation is more precise and likely exhibits less off-target effects.130 Recently, a study employing CRISPR technology to disrupt Maternal Embryonic Leucine Zipper Kinase (MELK) in vitro refuted the belief that MELK is essential for the viability of basal BC cells, despite continuing clinical trials investigating MELK inhibitors as chemotherapeutics. This study supports the necessity of employing CRISPR technology in preclinical target validation by challenging the justification for existing clinical trials.131 Motivated by the previously mentioned findings, they enhance the CRISPR/Cas9 system to specifically target the PARP1 gene to validate the selective synergism between PARP1 disruption and chemotherapy in TNBC cells. It evaluated several BRCA1 and PARP1 genetic profiles in both an in vitro 2D environment and a 3D tumor-on-a-chip system to more accurately replicate a physiological context.132

CAR-T cells

The most prevalent technique for generating tumor-specific T cells involves genetically altering T cells using CARs. Chimeric Antigen Receptors (CARs) typically comprise an extracellular ligand-binding domain derived from a single-chain antibody (scFv), a hinge region, a transmembrane domain, a cytoplasmic signaling chain, and/or costimulatory components. CAR-engineered T cells integrate the specificity of monoclonal antibodies with the homing and cytotoxic capabilities of T cells. CAR-T cell treatment is seen as having multiple advantages relative to other cellular immunotherapies. Initially, CAR-T cells are produced by nonspecifically activated polyclonal T cells. Consequently, they surmount the challenges associated with the isolation and amplification of natural tumor-specific CD4+ and CD8+ T cells.133,134 Secondly, CAR-T cells identify target antigens in a manner that is independent of MHC. This characteristic allows CAR-T cells to identify target cells with diminished HLA expression or antigen processing, which are regarded as significant contributors to tumor immunological evasion.135–137 Thirdly, CAR-T cells may actively and selectively migrate to tumor locations and can proliferate and endure long-term following tumor detection in vivo. Consequently, CAR-T cells directed against tumor-associated antigens (TAAs) may surpass monoclonal antibodies (mAbs) in eliciting enduring tumor responses.138

CAR-based therapy for solid tumors utilizes CARs aimed at colorectal cancer, ovarian cancer, prostate cancer, and metastatic renal cell carcinoma, among others.139 Research is also focused on HER-2, Lewis Y, mesothelin, folate receptor alpha (FR-α), and Muc1 for BC in vitro and in animal models.140,141 HER-2 expression is recognized to influence BC recurrence and overall survival. The application of anti-HER2 monoclonal antibodies has markedly enhanced BC prognosis. HER-2-targeted medicines are currently a fundamental aspect of the treatment for HER-2 overexpressing BC.142 Numerous clinical trials involving CAR-T cells aimed at HER-2 are currently underway, including a phase I/II study assessing HER-2-targeted CAR-T cells in conjunction with chemotherapy or HER-2 antibody inhibitor therapy for refractory HER-2 advanced BC, and a phase II study evaluating anti-CD3 x anti-HER2/Neu armed activated T cells following second-line chemotherapy in women with HER2/Neu (0, 1+ or 2+) metastatic BC.143

Conclusion

Only 5% of females experiencing breast discomfort are ultimately diagnosed with BC. The current BC treatment options encompass local therapies, systemic therapies and advanced systemic therapies. Due to advancements in screening, diagnosis, and treatment, the mortality rate from BC has decreased, with approximately 90% of newly diagnosed women surviving for at least five years. Ongoing research continues to improve the efficacy of screening and treatment methods. Future gene therapy candidates will be chosen based on tumor genetic data and the host's immune responses. With breakthroughs in safe gene delivery vectors and nuclease technology, genome editing holds promise as a novel approach for treating otherwise untreatable cancers. The trend in cancer treatment is shifting toward personalized care tailored to each patient's genome, immune system, and tumor profile. While CRISPR/Cas9 and CAR-T cell therapy show promise in stopping tumor growth, ethical questions, off-target consequences, and delivery challenges demand more study.

Funding

The authors have not received any funding for the study.

Ethical disclosures

It is a review article, and ethical disclosure is not applicable to this study.

References
[1]
R.L. Siegel, K.D. Miller, A. Jemal.
Cancer statistics, 2018.
CA Cancer J Clin, 68 (2018), pp. 7-30
[2]
K. Polyak.
Heterogeneity in breast cancer.
J Clin Invest, 121 (2011), pp. 3786-3788
[3]
P.C. Barathe, H.T. Haridas, P. Soni, K.K. Kudiya, J.B. Krishnan, V.S. Dhyani, et al.
Cost of breast cancer diagnosis and treatment in India: a scoping review protocol.
BMJ Open, 12 (2022),
[4]
G.N. Sharma, R. Dave, J. Sanadya, P. Sharma, K. Sharma.
Various types and management of breast cancer: an overview.
J Adv Pharm Technol Res, 1 (2010), pp. 109-126
[5]
A. Tassang, R.M. Brady, C.N. Fru, T. Yinka, T. Thierry, I.H. Musa, et al.
Breast cancer and prevention perspectives: a public health challenge, Buea Regional Hospital, Cameroon.
GSC Adv Res Rev, 13 (2022), pp. 258-268
[6]
U. Edward, E.I. Obeagu, H.M. Okorie, C.C. Vincent, Y.S. Bot.
Studies of serum calcium, inorganic phosphate and magnesium levels in lactating mothers in Owerri.
J Pharm Res Int, 33 (2021), pp. 209-216
[7]
E.I. Obeagu, Y.A. Ahmed, G.U. Obeagu, U.O. Bunu, O.P. Ugwu, E.U. Alum.
Biomarkers of breast cancer: overview.
Int J Curr Res Biol Med, 1 (2023), pp. 8-16
[8]
E.I. Obeagu, Q. Babar, C.C. Vincent, C.L. Udenze, R. Eze, C.J. Okafor, et al.
Therapeutic targets in breast cancer signaling: a review.
J Pharm Res Int, 33 (2021), pp. 82-99
[9]
Y.S. Sun, Z. Zhao, Z.N. Yang, F. Xu, H.J. Lu, Z.Y. Zhu, et al.
Risk factors and preventions of breast cancer.
Int J Biol Sci, 13 (2017), pp. 1387
[10]
C. Mattiuzzi, G. Lippi.
Current cancer epidemiology.
J Epidemiol Global Health, 9 (2019), pp. 217-222
[11]
Board PC.
BRCA1 and BRCA2: Cancer Risks and Management (PDQ®). InPDQ cancer information summaries.
National Cancer Institute (US), (2023),
[12]
S. Malvia, S.A. Bagadi, U.S. Dubey, S. Saxena.
Epidemiology of breast cancer in Indian women.
Asia Pac J Clin Oncol, 13 (2017), pp. 289-295
[13]
M.R. Monica.
An epidemiological study of cervical and breast screening in India: district-level analysis.
BMC Womens Health, 20 (2020), pp. 1
[14]
B. Chopra, V. Kaur, K. Singh, M. Verma, S. Singh, A. Singh.
Age shift: breast cancer is occurring in younger age groups-is it true.
Clin Cancer Investig J, 3 (2014),
[15]
G. Agarwal, P.V. Pradeep, V. Aggarwal, C.H. Yip, P.S. Cheung.
Spectrum of breast cancer in Asian women.
World J Surg, 31 (2007), pp. 1031-1040
[16]
Alkabban FM, Ferguson T. Breast cancer. StatPearls. Treasure Island (FL): StatPearls Publishing; 2020.
[17]
P.K. Dhillon.
Breast cancer fact sheet.
Mortality, 11 (2009), pp. 10-18
[18]
C.K. Anders, D.S. Hsu, G. Broadwater, C.R. Acharya, J.A. Foekens, Y. Zhang, et al.
Young age at diagnosis correlates with worse prognosis and defines a subset of breast cancers with shared patterns of gene expression.
J Clin Oncol, 26 (2008), pp. 3324-3330
[19]
S.P. Leong, Z.Z. Shen, T.J. Liu, G. Agarwal, T. Tajima, N.S. Paik, et al.
Is breast cancer the same disease in Asian and Western countries?.
World J Surg, 34 (2010), pp. 2308-2324
[20]
American College of Obstetricians and Gynecologists.
Hereditary cancer syndromes and risk assessment.
Obstet Gynecol, 134 (2019), pp. e143-e149
[21]
B. Xia, Q. Sheng, K. Nakanishi, A. Ohashi, J. Wu, N. Christ, et al.
Control of BRCA2 cellular and clinical functions by a nuclear partner, PALB2.
Mol Cell, 22 (2006), pp. 719-729
[22]
A. McTiernan.
Behavioral risk factors in breast cancer: can risk be modified?.
Oncologist, 8 (2003), pp. 326-334
[23]
Z. Abbas, S. Rehman.
An overview of cancer treatment modalities.
Neoplasm, 1 (2018), pp. 139-157
[24]
S. Charmsaz, D.M. Collins, A.S. Perry, M. Prencipe.
Novel strategies for cancer treatment: highlights from the 55th IACR annual conference.
Cancers (Basel), 11 (2019), pp. 1125
[25]
Z.S. Zumsteg, M. Morrow, B. Arnold, J. Zheng, Z. Zhang, M. Robson, et al.
Breast-conserving therapy achieves locoregional outcomes comparable to mastectomy in women with T1-2N0 triple-negative breast cancer.
Ann Surg Oncol, 20 (2013), pp. 3469-3476
[26]
M.C. van Maaren, L. de Munck, L.J. Strobbe, G.S. Sonke, P.J. Westenend, M.L. Smidt, et al.
Ten-year recurrence rates for breast cancer subtypes in The Netherlands: a large population-based study.
Int J Cancer, 144 (2019), pp. 263-272
[27]
E. Cipora, M. Konieczny, I.D. Karwat, W. Roczniak, M. Babuska-Roczniak.
Satisfaction with life among women with breast cancer-selected demographic and social factors.
Ann Agric Environ Med, 25 (2018),
[28]
S.P. Jackson, J. Bartek.
The DNA-damage response in human biology and disease.
Nature, 461 (2009), pp. 1071-1078
[29]
A.C. Begg, F.A. Stewart, C. Vens.
Strategies to improve radiotherapy with targeted drugs.
Nat Rev Cancer, 11 (2011), pp. 239-253
[30]
I. Mesa-Eguiagaray, S.H. Wild, P.S. Rosenberg, S.M. Bird, D.H. Brewster, P.S. Hall, et al.
Distinct temporal trends in breast cancer incidence from 1997 to 2016 by molecular subtypes: a population-based study of Scottish cancer registry data.
Br J Cancer, 123 (2020), pp. 852-859
[31]
M. Drãgãnescu, C. Carmocan.
Hormone therapy in breast cancer.
Chirurgia, 112 (2017), pp. 413-417
[32]
Early Breast Cancer Trialists' Collaborative Group.
Relevance of breast cancer hormone receptors and other factors to the efficacy of adjuvant tamoxifen: patient-level meta-analysis of randomised trials.
[33]
K.T. Flaherty, R.J. Gray, A.P. Chen, S. Li, L.M. McShane, D. Patton, et al.
Molecular landscape and actionable alterations in a genomically guided cancer clinical trial: National Cancer Institute Molecular Analysis for Therapy Choice (NCI-MATCH).
J Clin Oncol, 38 (2020), pp. 3883-3894
[34]
C. Sawyers.
Targeted cancer therapy.
Nature, 432 (2004), pp. 294-297
[35]
S. Dewanjee, J. Vallamkondu, R.S. Kalra, P. Chakraborty, M. Gangopadhyay, R. Sahu, et al.
The emerging role of HDACs: pathology and therapeutic targets in diabetes mellitus.
[36]
G. Mattheolabakis, B. Rigas, P.P. Constantinides.
Nanodelivery strategies in cancer chemotherapy: biological rationale and pharmaceutical perspectives.
Nanomedicine, 7 (2012), pp. 1577-1590
[37]
J.I. Leu, J. Pimkina, P. Pandey, M.E. Murphy, D.L. George.
HSP70 inhibition by the small-molecule 2-phenylethynesulfonamide impairs protein clearance pathways in tumor cells.
Mol Cancer Res, 9 (2011), pp. 936-947
[38]
W.K. Wu, Y.C. Wu, L. Yu, Z.J. Li, J.J. Sung, C.H. Cho.
Induction of autophagy by proteasome inhibitor is associated with proliferative arrest in colon cancer cells.
Biochem Biophys Res Commun, 374 (2008), pp. 258-263
[39]
International Breast Cancer Study Group (IBCSG).
Adjuvant chemotherapy followed by goserelin versus either modality alone for premenopausal lymph node–negative breast cancer: a randomized trial.
J Natl Cancer Inst, 95 (2003), pp. 1833-1846
[40]
M. Dowsett, J. Cuzick, J. Ingle, A. Coates, J. Forbes, J. Bliss, et al.
Meta-analysis of breast cancer outcomes in adjuvant trials of aromatase inhibitors versus tamoxifen.
J Clin Oncol, 28 (2010), pp. 509-518
[41]
C. Rose, O. Vtoraya, A. Pluzanska, N. Davidson, M. Gershanovich, R. Thomas, et al.
An open randomised trial of second-line endocrine therapy in advanced breast cancer: comparison of the aromatase inhibitors letrozole and anastrozole.
Eur J Cancer, 39 (2003), pp. 2318-2327
[42]
E.I. Buchbinder, A. Desai.
CTLA-4 and PD-1 pathways: similarities, differences, and implications of their inhibition.
Am J Clin Oncol, 39 (2016), pp. 98-106
[43]
J. Matsuzaki, S. Gnjatic, P. Mhawech-Fauceglia, A. Beck, A. Miller, T. Tsuji, et al.
Tumor-infiltrating NY-ESO-1–specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer.
Proc Natl Acad Sci, 107 (2010), pp. 7875-7880
[44]
F.M. Yakes, W. Chinratanalab, C.A. Ritter, W. King, S. Seelig, C.L. Arteaga.
Herceptin-induced inhibition of phosphatidylinositol-3 kinase and Akt Is required for antibody-mediated effects on p27, cyclin D1, and antitumor action.
Cancer Res, 62 (2002), pp. 4132-4141
[45]
S. Diermeier-Daucher, S. Breindl, S. Buchholz, O. Ortmann, G. Brockhoff.
Modular anti-EGFR and anti-Her2 targeting of SK-BR-3 and BT474 breast cancer cell lines in the presence of ErbB receptor-specific growth factors.
Cytometry A, 79 (2011), pp. 684-693
[46]
X.F. Le, F.X. Claret, A. Lammayot, L. Tian, D. Deshpande, R. LaPushin, et al.
The role of cyclin-dependent kinase inhibitor p27Kip1 in anti-HER2 antibody-induced G1 cell cycle arrest and tumor growth inhibition.
J Biol Chem, 278 (2003), pp. 23441-23450
[47]
R. Kumar, R. Yarmand-Bagheri.
The role of HER2 in angiogenesis.
Seminars in oncology, pp. 27-32
[48]
D.M. Pardoll.
The blockade of immune checkpoints in cancer immunotherapy.
Nat Rev Cancer, 12 (2012), pp. 252-264
[49]
M.E. Keir, S.C. Liang, I. Guleria, Y.E. Latchman, A. Qipo, L.A. Albacker, et al.
Tissue expression of PD-L1 mediates peripheral T cell tolerance.
J Exp Med, 203 (2006), pp. 883-895
[50]
L. Wang, V.S. Guillen, N. Sharma, K. Flessa, J. Min, K.E. Carlson, et al.
New class of selective estrogen receptor degraders (SERDs): expanding the toolbox of PROTAC degrons.
ACS Med Chem Lett, 9 (2018), pp. 803-808
[51]
X. Lin, H. Xiang, G. Luo.
Targeting estrogen receptor α for degradation with PROTACs: a promising approach to overcome endocrine resistance.
Eur J Med Chem, 206 (2020), pp. 112689
[52]
R.B. Kargbo.
PROTAC-mediated degradation of estrogen receptor in the treatment of cancer.
ACS Med Chem Lett, 10 (2019), pp. 1367-1369
[53]
H. Boulaiz, J.A. Marchal, J. Prados, C. Melguizo, A. Aranega.
Non-viral and viral vectors for gene therapy.
Cell Mol Biol (Noisy-le-Grand, France), 51 (2005), pp. 3-22
[54]
N.A. Kootstra, I.M. Verma.
Gene therapy with viral vectors.
Annu Rev Pharmacol Toxicol, 43 (2003), pp. 413-439
[55]
R. Fears, V. Ter Meulen.
Assessing security implications of genome editing: emerging points from an international workshop.
Front Bioeng Biotechnol, 6 (2018), pp. 34
[56]
J.E. Simón, Á.S. Rodríguez, Vispo N. Santiago.
CRISPR-Cas9: a precise approach to genome engineering.
Ther Innov Regul Sci, 52 (2018), pp. 701-707
[57]
K. Lundstrom.
Viral vectors in gene therapy.
[58]
J.M. Guedon, S. Wu, X. Zheng, C.C. Churchill, J.C. Glorioso, C.H. Liu, et al.
Current gene therapy using viral vectors for chronic pain.
Mol Pain, 11 (2015),
[59]
S. Ghosh, A.M. Brown, C. Jenkins, K. Campbell.
Viral vector systems for gene therapy: a comprehensive literature review of progress and biosafety challenges.
Appl Biosaf, 25 (2020), pp. 7-18
[60]
D.S. Strayer.
Viral vectors for gene therapy: past, present and future.
Drug News Perspect, 11 (1998), pp. 277-286
[61]
J.E. Chapman, D. Gillum, S. Kiani.
Approaches to reduce CRISPR off-target effects for safer genome editing.
Appl Biosaf, 22 (2017), pp. 7-13
[62]
J.E. Lunshof, A. Birnbaum.
Adaptive risk management of gene drive experiments: biosafety, biosecurity, and ethics.
Appl Biosaf, 22 (2017), pp. 97-103
[63]
Y.H. Chen, M.S. Keiser, B.L. Davidson.
Viral vectors for gene transfer.
Curr Protoc Mouse Biol, 8 (2018), pp. e58
[64]
S.L. Ginn, A.K. Amaya, I.E. Alexander, M. Edelstein, M.R. Abedi.
Gene therapy clinical trials worldwide to 2017: an update.
J Gene Med, 20 (2018), pp. e3015
[65]
J. Petrich, D. Marchese, C. Jenkins, M. Storey, J. Blind.
Gene replacement therapy: a primer for the health-system pharmacist.
J Pharm Pract, 33 (2020), pp. 846-855
[66]
C.S. Lee, E.S. Bishop, R. Zhang, X. Yu, E.M. Farina, S. Yan, et al.
Adenovirus-mediated gene delivery: potential applications for gene and cell-based therapies in the new era of personalized medicine.
[67]
M. Jafarlou, B. Baradaran, T.A. Saedi, V. Jafarlou, D. Shanehbandi, M. Maralani, et al.
An overview of the history, applications, advantages, disadvantages and prospects of gene therapy.
J Biol Regul Homeost Agents, 30 (2016), pp. 315-321
[68]
S.R. Soofiyani, B. Baradaran, F. Lotfipour, T. Kazemi, L. Mohammadnejad.
Gene therapy, early promises, subsequent problems, and recent breakthroughs.
Adv Pharm Bull, 3 (2013), pp. 249
[69]
S.M. Selkirk.
Gene therapy in clinical medicine.
Postgrad Med J, 80 (2004), pp. 560-570
[70]
Z. Karjoo, X. Chen, A. Hatefi.
Progress and problems with the use of suicide genes for targeted cancer therapy.
Adv Drug Deliv Rev, 99 (2016), pp. 113-128
[71]
M. Montaño-Samaniego, D.M. Bravo-Estupiñan, O. Méndez-Guerrero, E. Alarcón-Hernández, M. Ibáñez-Hernández.
Strategies for targeting gene therapy in cancer cells with tumor-specific promoters.
Front Oncol, 10 (2020), pp. 605380
[72]
F. Rossignoli, G. Grisendi, C. Spano, G. Golinelli, A. Recchia, G. Rovesti, et al.
Inducible Caspase9-mediated suicide gene for MSC-based cancer gene therapy.
Cancer Gene Ther, 26 (2019), pp. 11-16
[73]
F. Shafiee, M.G. Aucoin, A. Jahanian-Najafabadi.
Targeted diphtheria toxin-based therapy: a review article.
Front Microbiol, 10 (2019), pp. 2340
[74]
K. Higashi, S. Hazama, A. Araki, K. Yoshimura, N. Iizuka, S. Yoshino, et al.
A novel cancer vaccine strategy with combined IL-18 and HSV-TK gene therapy driven by the hTERT promoter in a murine colorectal cancer model.
Int J Oncol, 45 (2014), pp. 1412-1420
[75]
W.Y. Kuo, L. Hwu, C.Y. Wu, J.S. Lee, C.W. Chang, R.S. Liu.
STAT3/NF-κB-regulated lentiviral TK/GCV suicide gene therapy for cisplatin-resistant triple-negative breast cancer.
Theranostics, 7 (2017), pp. 647
[76]
G. Yang, X. Meng, L. Sun, N. Hu, S. Jiang, Y. Sheng, et al.
Antitumor effects of a dual cancer-specific oncolytic adenovirus on colorectal cancer in vitro and in vivo.
Exp Ther Med, 9 (2015), pp. 327-334
[77]
X. Zhou, M.K. Brenner.
Improving the safety of T-Cell therapies using an inducible caspase-9 gene.
Exp Hematol, 44 (2016), pp. 1013-1019
[78]
M. Mohseni-Dargah, S. Akbari-Birgani, Z. Madadi, F. Saghatchi, B. Kaboudin.
Carbon nanotube-delivered iC9 suicide gene therapy for killing breast cancer cells in vitro.
Nanomedicine, 14 (2019), pp. 1033-1047
[79]
V. Hoyos, F. Del Bufalo, S. Yagyu, M. Ando, G. Dotti, M. Suzuki, et al.
Mesenchymal stromal cells for linked delivery of oncolytic and apoptotic adenoviruses to non-small-cell lung cancers.
Mol Ther, 23 (2015), pp. 1497-1506
[80]
V. Pachnis, C.I. Brannan, S.M. Tilghman.
The structure and expression of a novel gene activated in early mouse embryogenesis.
[81]
I. Matouk, E. Raveh, P. Ohana, R.A. Lail, E. Gershtain, M. Gilon, et al.
The increasing complexity of the oncofetal h19 gene locus: functional dissection and therapeutic intervention.
Int J Mol Sci, 14 (2013), pp. 4298-4316
[82]
P. Ohana, I. Matouk, D. Amit, M. Gilon, A. Hochberg.
Toxin-based cancer gene therapy: under the control of oncofetal H19 regulatory sequences.
Gene Therapy Cancer, (2014), pp. 107-122
[83]
Y. Li, J. McCadden, F. Ferrer, M. Kruszewski, M. Carducci, J. Simons, et al.
Prostate-specific expression of the diphtheria toxin A chain (DT-A) studies of inducibility and specificity of expression of prostate-specific antigen promoter-driven DT-A adenoviral-mediated gene transfer.
Cancer Res, 62 (2002), pp. 2576-2582
[84]
H. Fung, S.L. Gerson.
Viral insertion site detection and analysis in cancer gene therapy.
Gene therapy of cancer, pp. 35-46
[85]
M. Gujrati, Z.R. Lu.
Targeted systemic delivery of therapeutic siRNA.
Gene therapy of cancer, pp. 47-65
[86]
Y. Sato, K. Murase, J. Kato, M. Kobune, T. Sato, Y. Kawano, et al.
Resolution of liver cirrhosis using vitamin A–coupled liposomes to deliver siRNA against a collagen-specific chaperone.
Nat Biotechnol, 26 (2008), pp. 431-442
[87]
M.E. Davis.
The first targeted delivery of siRNA in humans via a self-assembling, cyclodextrin polymer-based nanoparticle: from concept to clinic.
Mol Pharm, 6 (2009), pp. 659-668
[88]
J. Tabernero, G.I. Shapiro, P.M. LoRusso, A. Cervantes, G.K. Schwartz, G.J. Weiss, et al.
First-in-humans trial of an RNA interference therapeutic targeting VEGF and KSP in cancer patients with liver involvement.
Cancer Discov, 3 (2013), pp. 406-417
[89]
H. Li, Y. Yang, W. Hong, M. Huang, M. Wu, X. Zhao.
Applications of genome editing technology in the targeted therapy of human diseases: mechanisms, advances and prospects.
Signal Transduct Target Ther, 5 (2020), pp. 1
[90]
T.K. Guha, D.R. Edgell.
Applications of alternative nucleases in the age of CRISPR/Cas9.
Int J Mol Sci, 18 (2017), pp. 2565
[91]
J. Zhao, Q. Lin, Y. Song, D. Liu.
Universal CARs, universal T cells, and universal CAR T cells.
J Hematol Oncol, 11 (2018), pp. 1-9
[92]
H.X. Zhang, Y. Zhang, H. Yin.
Genome editing with mRNA encoding ZFN, TALEN, and Cas9.
Mol Ther, 27 (2019), pp. 735-746
[93]
H. Bi, B. Yang.
Gene editing with TALEN and CRISPR/Cas in rice.
Prog Mol Biol Transl Sci, 149 (2017), pp. 81-98
[94]
W. Ding, Z. Hu, D. Zhu, X. Jiang, L. Yu, X. Wang, et al.
Zinc finger nucleases targeting the human papillomavirus E7 oncogene induce E7 disruption and a transformed phenotype in HPV16/18-positive cervical cancer cells.
Clin Cancer Res, 20 (2014), pp. 6495-6503
[95]
S. Shankar, D. Prasad, R. Sanawar, A.V. Das, M.R. Pillai.
TALEN based HPV-E7 editing triggers necrotic cell death in cervical cancer cells.
[96]
R. Puria, S. Sahi, V. Nain.
HER2+ breast cancer therapy: by CPP-ZFN mediated targeting of mTOR?.
Technol Cancer Res Treat, 11 (2012), pp. 175-180
[97]
C. Ashmore-Harris, G.O. Fruhwirth.
The clinical potential of gene editing as a tool to engineer cell-based therapeutics.
Clin Transl Med, 9 (2020), pp. 15
[98]
F.A. Khan, N.S. Pandupuspitasari, H. Chun-Jie, Z. Ao, M. Jamal, A. Zohaib, et al.
CRISPR/Cas9 therapeutics: a cure for cancer and other genetic diseases.
Oncotarget, 7 (2016), pp. 52541
[99]
X. Zhao, L. Liu, J. Lang, K. Cheng, Y. Wang, X. Li, et al.
A CRISPR-Cas13a system for efficient and specific therapeutic targeting of mutant KRAS for pancreatic cancer treatment.
Cancer Lett, 431 (2018), pp. 171-181
[100]
N. Kawamura, K. Nimura, H. Nagano, S. Yamaguchi, N. Nonomura, Y. Kaneda.
CRISPR/Cas9-mediated gene knockout of NANOG and NANOGP8 decreases the malignant potential of prostate cancer cells.
Oncotarget, 6 (2015), pp. 22361
[101]
W. Li, M.Y. Cho, S. Lee, M. Jang, J. Park, R. Park.
CRISPR-Cas9 mediated CD133 knockout inhibits colon cancer invasion through reduced epithelial-mesenchymal transition.
PLoS ONE, 14 (2019), pp. e0220860
[102]
G. Domenici, I. Aurrekoetxea-Rodríguez, B.M. Simões, M. Rábano, S.Y. Lee, J.S. Millán, et al.
A Sox2–Sox9 signaling axis maintains human breast luminal progenitor and breast cancer stem cells.
Oncogene, 38 (2019), pp. 3151-3169
[103]
B.N. Hannafon, A. Cai, C.L. Calloway, Y.F. Xu, R. Zhang, K.M. Fung, et al.
MiR-23b and miR-27b are oncogenic microRNAs in breast cancer: evidence from a CRISPR/Cas9 deletion study.
BMC Cancer, 19 (2019), pp. 642
[104]
T. Zhan, N. Rindtorff, J. Betge, M.P. Ebert, M. Boutros.
CRISPR/Cas9 for cancer research and therapy.
Semin Cancer Biol, 55 (2019), pp. 106-119
[105]
S. Patil, Y.G. Gao, X. Lin, Y. Li, K. Dang, Y. Tian, et al.
The development of functional non-viral vectors for gene delivery.
Int J Mol Sci, 20 (2019), pp. 1-23
[106]
B. Dhungel, A. Jayachandran, C.J. Layton, J.C. Steel.
Seek and destroy: targeted adeno-associated viruses for gene delivery to hepatocellular carcinoma.
Drug Deliv, 24 (2017), pp. 289-299
[107]
S. Hao, X. Du, Y. Song, M. Ren, Q. Yang, A. Wang, et al.
Targeted gene therapy of the HSV-TK/hIL-12 fusion gene controlled by the hSLPI gene promoter of human non-small cell lung cancer in vitro.
Oncol Lett, 15 (2018), pp. 6503-6512
[108]
R. Mohammadinejad, A. Dehshahri, V.S. Madamsetty, M. Zahmatkeshan, S. Tavakol, P. Makvandi, et al.
In vivo gene delivery mediated by non-viral vectors for cancer therapy.
J Control Release, 325 (2020), pp. 249-275
[109]
P. Zhang, J. Tan, D.B. Yang, Z.C. Luo, S. Luo, P. Chen, et al.
Gene therapy using the human telomerase catalytic subunit gene promoter enables targeting of the therapeutic effects of vesicular stomatitis virus matrix protein against human lung adenocarcinoma.
Exp Ther Med, 4 (2012), pp. 859-864
[110]
I.V. Alekseenko, V.V. Pleshkan, A.V. Sass, O.B. Filyukova, E.V. Snezhkov, E.D. Sverdlov.
A universal tumor-specific promoter for cancer gene therapy.
Doklady biochemistry and biophysics, pp. 158-161
[111]
C. Chen, D. Yue, L. Lei, H. Wang, J. Lu, Y. Zhou, et al.
Promoter-operating targeted expression of gene therapy in cancer: current stage and prospect.
Mol Therapy-Nucl Acids, 11 (2018), pp. 508-514
[112]
Y. Ishino, M. Krupovic, P. Forterre.
J Bacteriol, 200 (2018), pp. 10
[113]
M. Jinek, K. Chylinski, I. Fonfara, M. Hauer, J.A. Doudna, E. Charpentier.
Science, 337 (2012), pp. 816
[114]
O. Shalem, N.E. Sanjana, F. Zhang.
Nat Rev Genet, 16 (2015), pp. 299
[115]
J. Pulecio, N. Verma, E. Mejía-Ramírez, D. Huangfu, A. Raya.
CRISPR/Cas9-based engineering of the epigenome.
Cell Stem Cell, 21 (2017), pp. 431-447
[116]
D. Bhaya, M. Davison, R. Barrangou.
CRISPR-Cas systems in bacteria and archaea: versatile small RNAs for adaptive defense and regulation.
Annu Rev Genet, 45 (2011), pp. 273-297
[117]
I. Yosef, M.G. Goren, U. Qimron.
Proteins and DNA elements essential for the CRISPR adaptation process in Escherichia coli.
Nucleic Acids Res, 40 (2012), pp. 5569-5576
[118]
J.K. Nuñez, P.J. Kranzusch, J. Noeske, A.V. Wright, C.W. Davies, J.A. Doudna.
Cas1–Cas2 complex formation mediates spacer acquisition during CRISPR–Cas adaptive immunity.
Nat Struct Mol Biol, 21 (2014), pp. 528-534
[119]
R. Heler, P. Samai, J.W. Modell, C. Weiner, G.W. Goldberg, D. Bikard, et al.
Cas9 specifies functional viral targets during CRISPR–Cas adaptation.
Nature, 519 (2015), pp. 199-202
[120]
E. Deltcheva, K. Chylinski, C.M. Sharma, K. Gonzales, Y. Chao, Z.A. Pirzada, et al.
CRISPR RNA maturation by trans-encoded small RNA and host factor RNase III.
Nature, 471 (2011), pp. 602-607
[121]
G. Gasiunas, R. Barrangou, P. Horvath, V. Siksnys.
Cas9–crRNA ribonucleoprotein complex mediates specific DNA cleavage for adaptive immunity in bacteria.
Proc Natl Acad Sci, 109 (2012), pp. E2579-E2586
[122]
C. Anders, O. Niewoehner, A. Duerst, M. Jinek.
Structural basis of PAM-dependent target DNA recognition by the Cas9 endonuclease.
Nature, 513 (2014), pp. 569-573
[123]
M. Jinek, F. Jiang, D.W. Taylor, S.H. Sternberg, E. Kaya, E. Ma, et al.
Structures of Cas9 endonucleases reveal RNA-mediated conformational activation.
Science, 343 (2014), pp. 1247997
[124]
D.R. Rodríguez-Rodríguez, R. Ramirez-Solis, M.A. Garza-Elizondo, M. de Lourdes Garza-Rodriguez, H.A. Barrera-Saldana.
Genome editing: a perspective on the application of CRISPR/Cas9 to study human diseases.
Int J Mol Med, 43 (2019), pp. 1559-1574
[125]
B.A. Lone, S.K. Karna, F. Ahmad, N. Shahi, Y.R. Pokharel.
CRISPR/Cas9 system: a bacterial tailor for genomic engineering.
Genet Res Int, 2018 (2018),
[126]
J.H. Hu, S.M. Miller, M.H. Geurts, W. Tang, L. Chen, N. Sun, et al.
Evolved Cas9 variants with broad PAM compatibility and high DNA specificity.
Nature, 556 (2018), pp. 57-63
[127]
M. Chehelgerdi, M. Chehelgerdi, M. Khorramian-Ghahfarokhi, M. Shafieizadeh, E. Mahmoudi, F. Eskandari, et al.
Comprehensive review of CRISPR-based gene editing: mechanisms, challenges, and applications in cancer therapy.
[128]
L.A. Gilbert, M.H. Larson, L. Morsut, Z. Liu, G.A. Brar, S.E. Torres, et al.
CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes.
[129]
A. Chavez, J. Scheiman, S. Vora, B.W. Pruitt, M. Tuttle, E.P.R. Iyer, et al.
Highly efficient Cas9-mediated transcriptional programming.
Nat Methods, 12 (2015), pp. 326-328
[130]
R.L. Mintz, M.A. Gao, K. Lo, Y.H. Lao, M. Li, K.W. Leong.
CRISPR technology for breast cancer: diagnostics, modeling, and therapy.
Adv Biosyst, 2 (2018), pp. 1800132
[131]
S. Konermann, P. Lotfy, N.J. Brideau, J. Oki, M.N. Shokhirev, P.D. Hsu.
Transcriptome engineering with RNA-targeting type VI-D CRISPR effectors.
[132]
A. Lin, C.J. Giuliano, N.M. Sayles, J.M. Sheltzer.
CRISPR/Cas9 mutagenesis invalidates a putative cancer dependency targeted in on-going clinical trials.
Elife, 6 (2017), pp. e24179
[133]
F. Le Du, B.L. Eckhardt, B. Lim, J.K. Litton, S. Moulder, F. Meric-Bernstam, et al.
Is the future of personalized therapy in triple-negative breast cancer based on molecular subtype?.
Oncotarget, 6 (2015), pp. 12890
[134]
M. Kanematsu, M. Futamura, M. Takata, S. Gaowa, A. Yamada, K. Morimitsu, et al.
Clinical significance of glycoprotein nonmetastatic B and its association with HER2 in breast cancer.
Cancer Med, 4 (2015), pp. 1344-1355
[135]
F.C. Geyer, F. Pareja, B. Weigelt, E. Rakha, I.O. Ellis, S.J. Schnitt, et al.
The spectrum of triple-negative breast disease: high-and low-grade lesions.
Am J Pathol, 187 (2017), pp. 2139-2151
[136]
H. Yao, G. He, S. Yan, C. Chen, L. Song, T.J. Rosol, et al.
Triple-negative breast cancer: is there a treatment on the horizon?.
Oncotarget, 8 (2017), pp. 1913-1924
[137]
A. Prat, A. Lluch, J. Albanell, W.T. Barry, C. Fan, J.I. Chacón, et al.
Predicting response and survival in chemotherapy-treated triple-negative breast cancer.
Br J Cancer, 111 (2014), pp. 1532-1541
[138]
J. Yang, J. Nie, X. Ma, Y. Wei, Y. Peng, X. Wei.
Targeting PI3K in cancer: mechanisms and advances in clinical trials.
[139]
H. Sun, A.H. Zhang, S.B. Liu, S. Qiu, X.N. Li, T.L. Zhang, et al.
Cell metabolomics identify regulatory pathways and targets of magnoline against prostate cancer.
J Chromatogr B, 1102 (2018), pp. 143-151
[140]
N.R. Datta, S.G. Ordóñez, U.S. Gaipl, M.M. Paulides, H. Crezee, J. Gellermann, et al.
Local hyperthermia combined with radiotherapy and−/or chemotherapy: recent advances and promises for the future.
Cancer Treat Rev, 41 (2015), pp. 742-753
[141]
P. Frit, N. Barboule, Y. Yuan, D. Gomez, P. Calsou.
Alternative end-joining pathway (s): bricolage at DNA breaks.
DNA Repair, 17 (2014), pp. 81-97
[142]
A.L. Oei, H.P. Kok, S.B. Oei, M.R. Horsman, L.J. Stalpers, N.A. Franken, et al.
Molecular and biological rationale of hyperthermia as radio-and chemosensitizer.
Adv Drug Deliv Rev, 163 (2020), pp. 84-97
[143]
X. Wu, A.J. Kriz, P.A. Sharp.
Target specificity of the CRISPR-Cas9 system.
Quantit Biol, 2 (2014), pp. 59-70
Descargar PDF
Opciones de artículo
es en pt

¿Es usted profesional sanitario apto para prescribir o dispensar medicamentos?

Are you a health professional able to prescribe or dispense drugs?

Você é um profissional de saúde habilitado a prescrever ou dispensar medicamentos