Au nanoparticles in the diagnosis and treatment of ovarian cancer: A new horizon in the personalized medicine

Document Type : Review Paper

Authors

1 Department of Surgery, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran

2 Obstetrics and Gynecology specialist, faculty of medicine, Kermanshah university of medical science, Kermanshah, Iran

3 Department of Surgery, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran

4 Sinop University, Faculty of Health Science, Sinop, Turkey

5 Obstetric and Gynecologist Specialist, Isfahan University of Medical Sciences, Isfahan, Iran

Abstract

As the world's sixth prevalent malignancy among women, the increased rate of mortality in ovarian cancer (OC)  patients is due to late diagnosis that causes a high rate of proliferation within the abdominal cavity. The sensitivity of screening and detection methods for the diagnosis of ovarian cancer in the early stages is insufficient. Considering the high rate of ovarian cancers resistance to most traditional treatments that cause the risk of disease recurrence and death, it is necessary to design new treatments and diagnostic methods. In this regard, nanoparticles and nanotechnology can be viable options for suppressing these limitations. One of the goals of nanotechnology is to improve the approaches of diagnosing, treating, or their combination (theranostics) in a variety of diseases including cancer. Au nanoparticles can simultaneously integrate therapeutic and imaging agents due to their special and extraordinary physicochemical properties and function as theranostic platforms. Next to their numerous distinct features, such as small size, surface impacts, quantum size, and electrical and optical effects, AuNPs proved to be relatively secure, stable, and require a simple preparation. Gold nanoparticles can be exerted as carriers for a more effective and targeted diagnostic and therapeutic agent delivery in the treatment of ovarian cancer. They can limit drug toxicity at tumor site and consequently reduce the toxicity of normal cells and tissues. Gold nanoparticles can be used as nano-theranostics agents and facilitate personalized medicine for a more efficient treatment of ovarian cancer by providing the simultaneous delivering of diagnostic and therapeutic agents.

Keywords


INTRODUCTION
Ovarian cancer (OC) is the world’s sixth prevalent malignancy in women that leads to the death of more patients than the other malignancies of female reproductive system. [1]. Ovarian cancer cells are a heterogeneous group of cancer cells. Malignant tumour cells primarily disseminate in the peritoneal cavity and tend to multiply rapidly, putting pressure on the visceral organs. Ovarian carcinoma is a particularly dangerous illness with a low cure rate of 30% that only temporarily respond to chemotherapy. Many genetic and epigenetic factors can contribute to the transformation of ovarian cell carcinoma cells. With the ability to quickly grow and proliferate, the delayed diagnosis of these cells can induce ascites, intestinal obstruction, and tumour cachexia. Certain factors can reduce the danger of ovarian cancer in different patients such as increasing the number of alive births, enlarging the number of unfinished pregnancies, and using oral contraceptives. Women that suffer from obesity, cervical polyps, and gallbladder illnesses, as well as those with a “immediate” intolerance to contraceptives, are all linked to an elevated chance of developing this disease [2-5]. There are numerous studies with solid proofs on the significant effects of age on the progress of ovarian cancer. According to these researches, women in the age of 65 and above have a higher chance of containing ovarian cancer than the younger individuals . Also, a significant increase was reported in the mortality rate of this cancer in women aged 65 and above [6]. According to the data of BRCA1 / 2 gene mutations, there are similar biological features among the basal-like breast tumors and ovarian malignancies that include containing a high level of genomic instability and being incapable of repairing DNA. There are factors that can contribute to this cancer including deficient pregnancies, specific surgeries like hysterectomy and tubal ligation, related family history, oophorectomy, oral contraceptive use, and postmenopausal hormone replacement therapy, as well as lifestyle habits such as smoking and consuming alcohol [7, 8]. The majority of ovarian cancer patients are diagnosed when facing the advanced levels of this disease. The combination of surgical debulking and drug treatment, as well as chemotherapy and  radiation therapy, are among the list of currently available therapeutic options for front-line therapy . Although a high percentage of advanced ovarian cancer patients respond to the first treatment, yet 70% of the cases relapse to their disease and ultimately face death [9]. The high rate of ovarian cancer resistance to most chemotherapy drugs points out the importance of discovering novel treatments and emphasizes the need for significant advances in therapeutic strategies and early detection. The advances of nanotechnology provided the design of related approaches that can be adapted for the treatment of chemo-resistant OC. Nano-based therapeutics can function as enticing viable alternatives for the conventional therapies of various diseases including malignancies [10]. The outcomes of patients’ treatments can be improved by the application of Nanoparticles (NPs) as an applicable platform for delivering cytotoxic agents and facilitating early disease diagnosis. The biological and photothermal therapeutic applications of Au nanoparticles (AuNPs) were extensively investigated in recent years. AuNPs are capable of binding to a variety of organic compounds while containing a simple synthesizing procedure [11]. This article provided a summary on the properties of AuNPs, their contribution to tumor diagnosis, and their potential for treating ovarian cancer. In this regard, we attempted to study gold nanoparticles, which stand as a new horizon in the field of personalized medicine.

OVARIAN CANCER (OC) SCREENING TEST 
The patients of ovarian cancer commonly show a low rate of endurance due to their late-stage detection and diagnosis. On the other hand, public screening is neither cost-effective nor practicable. However, there are specific patient subgroups that volunteer for ovarian cancer screening (mostly those with genetic risk factors). Bimanual pelvic examination, cancer antigen (CA) 125, and transvaginal ultrasonography are among the available screening procedures in current years. Pelvic examination can be an inexpensive treatment for women who are already on  regular schedule of taking gynecologic exams and their results can be accountable if performed by a trained examiner; however, this route lacks the required sensitivity and specificity as a screening test [12, 13]. 80 percent of ovarian carcinomas cases report an elevation in the radioimmunoassay of CA 125, which is a tumor-specific antigen, whereas this inducement is observed in only 50 percent of women with ovarian cancer. The elevated state of this factor is also observed in women with benign ovarian illness and those who are generally healthy, which limits its specificity. Transvaginal ultrasound (TVUS) is one of the available screening methods for OC, which implicates transvaginal ultrasonography for identifying the growth and masses throughout the scanned area. Next to being quiet costly, ultrasound is incapable of providing a sufficient sensitivity and specificity for distinguishing the cases of malignant and benign tumors. Therefore, exerting a combination of TVUS and CA125 levels in patients with high CA125 levels may be a better screening strategy for the diagnosis of OC [14-16].

DIAGNOSIS OF OVARIAN CANCER
Ovarian cancer is recognized as the leading cause of gynecological cancer death due to displaying few early symptoms, being noticeable at an advanced stage, and containing poor chances of survival. The related researches of last decade attempted to focus on improving the outcomes of ovarian cancer treatments by utilizing imaging techniques and serum indicators to screen the early stage of this disease for preclinical purposes. The common EOC symptoms that appear months before diagnosis include abdominal bloating, early satiety, nausea, abdominal distension, alteration in bowel function, urinary symptoms, back pain, fatigue, and weight loss. The measurement of CA125 concentrations and the performance of pelvic ultrasound are included throughout the initial examination. The conduction of further imaging, such as chest, abdomen, or pelvis CTs for staging, as well as pelvic MRI if required, should be considered to correctly characterize the extension of EOC. Furthermore, it is crucial to have a pathological detection on tumor tissue due to the varying histological subgroups of ovarian cancer that implicate different treatment methods. In conformity to the reported observations over the last decade, EOC is known as a collection of disorders with unique precursor lesions, tissues of origin, molecular biology, clinical presentation, chemosensitivity, and patient outcome [17-20]. The increased mortality in OVC patients is due to the high rate of death caused by its late diagnosis, which leads to a massive proliferation throughout the abdominal cavity. The early identification of OVCA can be facilitated through the technological advancements that implicate physical diagnosis, clinical histories, CA-125 serum protein detection, physical evaluation, and ultrasound examination[21-23].

COMMON TREATMENTS OF OVARIAN CANCER
Standard stages of ovarian cancer management:
The typical treatment of advanced ovarian cancer involves a primary cytoreductive surgery that is followed by performing platinum-based chemotherapy. There are two types of ovarian cancer surgeries that include simple surgery and radical surgery, both of which are parts of primary cytoreductive surgery. Achieving a better comprehension of this disease and its natural history has also enlightened the concept of staging surgery in the beginning levels. Cytoreductive surgery is performed to accurately establish a diagnosis, remove poor perfusion, and reduce the tumor size for the purpose of improving the adjuvant chemotherapy[24, 25].  The majority of patients are subjected to simple surgery that is comprised of hysterectomy and bilateral salpingooophorectomy, infracolic omentectomy, limited retroperitoneal node excision, and segmental resection of small intestine, while involving the minimum risk of complications. The more elaborated treatments of radical surgery implicate a higher risk of bleeding and complications, as well as a longer surgery time and hospital stay, which can be reduced by proper preoperative and postoperative care [26, 27]. The data of tomographic findings from related studies helped in predicting the inability of adequate cytoreduction due to certain factors such as massive ascites, hepatic parenchyma metastases, severe diaphragmatic illness, and other abnormalities. On the other hand, the performance of laparoscopy provides a direct line of sight into the disease’s progression. Nevertheless, the patient must consider many parameters before choosing between surgery and neoadjuvant chemotherapy [28, 29]. The probability of metastasis in various areas of peritoneal cavity was estimated to reach up to 11 percent. In addition, the possible occurrence of metastasis throughout the omentum was estimated to be 35 percent, while the chance of having malignant cells in peritoneal lavage was predicted to be 33 percent. Other possible sites of metastasis include lymph node, specifically in the aortic node, with a 2–24 percent probable chance of occurrence  throughout the beginning stages of the disease, while the inducement of metastases in pelvic iliac nodes were observed in up to 8–15 percent of the cases [27, 30, 31]. The most common drugs used for the chemotherapy of ovarian cancer include carboplatin, paclitaxel, cisplatin, platinum, doxorubicin (DOX), decitabine (DB), and gemcitabine. According to outcomes, surgery can be a desirable method for tumors that are sized in less than 1 cm. Nevertheless, 75% of patients notice their disease at its advanced (stage III or IV) levels and despite benefiting from first-line therapy, their tumor recurrence occurs in about 15 months from diagnosis [8, 32-35]. Many trials attempted to increase clinical responses (CR) and reduce the induced toxicity in advanced ovarian cancer patients through different chemotherapeutic drugs and combinations [36]. Table 1 provides an overview of the exerted chemotherapy regimens in the treatment of advanced ovarian cancer.

The most important disadvantages of chemotherapy
In general, the overall survival and progression ‐ free survival of patients with advanced ovarian cancer can be increased through chemotherapy [52-54]. The exerted drugs in chemotherapy, including alopecia, gastrointestinal effects ,infection myelosuppression and neuropathy, are associated with unwanted side effects [53, 55] and may also implicate the risk of causing drug resistance. Drug resistance can occur for a variety of reasons, including pharmacokinetic, micro-environmental tumor, and cancer-cell-specific abnormalities, which may lead to death in some cases [56]. Multidrug resistance (MDR) stands as a critical barriers to an effective and comprehensive ovarian cancer treatment. The proposed hypotheses for determining the actual underlying causes of MDR and disease relapse are listed in the following [57]. Changes in drug efflux: As an eminent cause of MDR advancement, the direct efflux of chemotherapeutic drugs into the extracellular environment of cells is mediated by the ATP-binding cassette (ABC) family and ATP hydrolysis. The ABC family of transporters with 49 members is responsible for maintaining the intracellular drug concentrations [58, 59].
DNA damage repair dysfunction: A damaged DNA structure by physical, chemical, or biological factors results in triggering the functionality of DNA repair pathway and repair genes to preserve the genome’s integrity and stability. On the other hand, the ectopic activation of DNA repair mechanism leads to the frequent progression of tumors. Since the mechanism of chemotherapeutics implicates direct or indirect processes to induce DNA damage and consequently annihilate the tumor cells, the repair abilities of tumor cells’ DNA are intensified and eventually lead to the development of MDR[60, 61]. 
Apoptosis: There are two principal apoptotic processes that include extrinsic or death receptor pathway that is balanced by tumor necrosis factor (TNF) receptors, and the intrinsic or mitochondrial pathway that is accorded by chondriosomes. This balance can be disrupted by the Bcl-2 protein family, which is consisted of pro-apoptotic proteins such as Bax, Bad, and Bid along with anti-apoptotic proteins such as Bcl-2 and Bcl-xl.Any. This inducement  can be considered as a major factor in extending chemotherapeutic resistance and even lead to apoptotic inhibition during carcinogenesis[62, 63].
Drug target mutation: Molecular targeted treatment is recognized as a progressive therapeutic strategy for the cases of cancer. Numerous studies were conducted on this topic due to its lower adverse impacts and higher efficacy than traditional chemotherapy agents., Among the other chemicals and proteins, this technique can implicate the members of signaling pathway in the role of potential targets; nevertheless, other studies reported the possible development of MDR upon the occurrence of a signaling system alteration[58, 64].
Epigenetic alterations:  As the principal cause of carcinogenesis, Epigenetic alterations is a unique and crucial method in the development of MDR. The two basic types of epigenetic alterations include DNA methylation, which facilitates the binding of methyl groups to cytosines in CpG islands, and histone modification induced by acetylation or methylation. Several studies confirmed the role of epigenetic alterations, including histone methylation and acetylation and DNA methylation, in the occurrence of cancer cell resistance to different chemotherapeutic drugs [65, 66]. Up to this date, all of the conducted therapies ended in a significant rate of relapse and unsatisfying treatment outcomes, which point out the necessity of performing further researches to discover new therapeutic regimens for ovarian cancer (OVCA)  patients [67].

NANOTECHNOLOGY-BASED TECHNIQUES: A POTENT SOLUTION TO SURPASS THE LIMITS OF TRADITIONAL METHODS IN THE DIAGNOSIS AND TREATMENT OF OVARIAN CANCER
Ovarian carcinoma stands as the leading reason of death by gynecologic malignancy, which is caused by delayed initial detection and the recurrence of ovarian cancer (OVCA) due to therapeutic resistance. Many patients experience OVCA recurrence, which makes them resistant to treatment and incurable. According to updated reports, recent observations on ovarian cancer stem cells (CSCs) indicated their high resistance towards conventional cytotoxic chemotherapeutic drugs and confirmed their potent contribution to treatment resistance and OVCA recurrence. The current diagnostic and therapy methods lack the sufficient sensitivity or efficacy for diagnosing OVCA in its early stages, while the high expenditures and absence of a specified detection point results in a late diagnosis. Nanotechnology has proven its strong applicability in the diagnosis and treatment of OVCA while facing an increasing rate of application in this field. [68-70]. The goal of this technology is to enhance the available methods of diagnosing, treating, or their combination  (theranostics) for a variety of diseases such as cancer. The exertion of nanocarriers can facilitate the targeted delivery of hydrophobic chemicals, delivery of carrier stabilization, and reduce the systemic toxicity of antineoplastic agent, as well as API biodistribution and pharmacokinetics. The usage of physical adsorption or chemical conjugations can provide the loading of nanocariers with drugs, imaging agents, targeting moieties such as ligands or antibodies, and polyethylene glycol (PEG) in order to extend the half-life of therapeutic agents and improve passive and active tumor targeting [10, 71, 72]. Liposomes, self-assembled polymers, micelles, hydrogels, dendrimers, quantum dots, magnetic nanoparticles, carbon-based nanocarriers (bucky balls and carbon nanotubes), and metal or oxide based nanoparticles (colloidal Au, silica, and titanium dioxide) are some of exerted nanocarriers for the diagnosis and treatment of various types of cancers, especially ovarian cancer. Due to their exceptional physicochemical and optical features, Au nanoparticles were designated in the form of cutting-edge platforms to combine diagnostic and therapeutic strategies. These products may provide significant benefits in cancer settings by taking the role of targeted vehicles to control the drug release, photothermal therapy, and gene therapy, as well as functioning in the form of contrast imaging agents to monitor the disease and therapeutic progress in real time. These image-guided therapeutic platforms can be an effective treatment with an adjustable design  for maximizing the personal benefits of patients while minimizing the hazardous side effects[73, 74].

Au nanoparticles
Au is a material with high thermal and electrical conductivity, which can act as an excellent reflector of IR infrared waves. Some of the most important properties of AuNPs at nanoscale include the increased cross section of materials due to their smaller particle size, intensified reactivity[75, 76] and low toxicity[77], as well as being eco-friendly[78] and stable[79] next to containing surface plasma resonance[80], the option of surface modification and functionalization[81], and high thermal[82] and electrical conductivity[83]. Furthermore, these nanoparticles have a wide range of applications and medical implementations due to their identical biocompatibility qualities. They also contain an electronic-optical structure that led to the rise of several applications in the fields of sensors and biosensors. AuNPs can be used as an antibacterial agent[84], the release of drugs and therapeutic agents[85] in nanowires and catalytic applications[86], Hyperthermia[87], and sensors[88], as well as for the fabrication of electronic chips[89], Raman spectroscope[90] and Diagnosis of diseases [91]. The unique factors of these products has characterized them in a different class from conventional cancer treatments. Their simple synthesis in various sizes and shapes provided a variety of optical properties including intense light absorption and scattering, a high rate of photothermal conversion, photostability, and colloidal stability, as well as being biocompatible, contain a simple ligand conjugation chemistry, and facilitate extended molecule loading per particle. Due to their reduced particle size and surface chemistry, Au nanoparticles can be coated with small molecules, aptamers, polymers, and biomarkers in order to expand their range of applications [92-94]. Next to their ability to carry active chemicals, AuNPs stand as potent theranostic platforms since they can exhibit a high affinity and specificity for cancer cells as a result of containing various targeting moieties, while simultaneously holding therapeutic and imaging agents [92-95]. 

AuNPs in the diagnosis of ovarian cancer: imaging-based detection
Although there is a lack of any available routine screening of OVCA for healthy women, yet there are some screening options for at-risk patients that implicate a routine pelvic examination, transvaginal sonography, and a blood serum biomarker test known as CA125. However, the current screening and detection methods lack the sufficient sensitivity for identifying OVCA in its early stages in order to improve the patient’s prognosis by treatment interference. In addition, exorbitant expenditures and a lack of point-of-care screening procedures lead to a delayed diagnosis and inadequate care for the patients. Biomarker screening provides the best results upon being performed on blood, urine, or saliva samples in people at the risk of developing OVCA [96, 97]. The accommodation of considerably improved radiative features, electromagnetic field improvement, and the ability to conjugate with contrast agents proves the effectiveness of AuNPs in the role of imaging agents for the in vitro and in vivo imaging-based diagnostics of cancer cells (Table 2). Light scattering imaging, two-photon fluorescence (TPF) imaging, photoacoustic imaging / tomography (PAT), x-ray computed tomography (CT), surface-enhanced Raman spectroscopy (SERS), and magnetic resonance imaging (MRI) can be exerted in the course of this procedure[98-100].

AuNPs in the treatment of ovarian cancer
Although studies have confirmed the effects of performing treatment with conventional chemotherapy and surgery in improving the statues of advanced ovarian cancer patients, however, most of the cases end with death due to drug-resistant subsequent to 5 years. Therefore, the enhancement of molecular targeted therapies may lead to the design of more effective treatments [119]. The ineffectiveness of traditional therapies in treating ovarian cancer is undeniable, which highlights the necessity of designing novel approaches to improve the obtained efficacy [120]. In fact, the exertion of various chemotherapy agents can enhance the survival rate of patients with ovarian cancer, but their application is limited by their distribution through the whole body, which leads to high normal organ toxicity. The small sizes (10-100 nm) of particles in nanotechnology can improve blood circulation and enhance the status of accumulated therapeutic drugs at the tumor sites [121, 122]. In addition, another fundamental barrier in the treatment of ovarian cancer is drug resistance, which can be most probably surpassed through the provided approaches by nanotechnology. The formulations that are based upon nanotechnology  involve encapsulated, conjugated, or entrapped / loaded forms in nanocarriers or drug delivery vehicle / vectors [123, 124]. The controlled delivery of chemotherapeutic drug (s) in a targeted attempt can lead to the achievement of nanotechnology-based therapies that would directly affect the cancer site for extended time intervals while causing a minimal or zero normal organ toxicity[125]. The administration of numerous traditional chemotherapeutic agents are conducted through oral or intravenous routes, which contain weak pharmacokinetics along with a narrow therapeutic window. Next to their quick rate of reaching the maximum tolerated concentration in the blood, these drugs leave the bloodstream afterwards due to their low blood circulation half-life[126, 127]. The side effects of anti-cancer drug, similar to the impacts of radiotherapy, remain a major limitation in the advancement of cancer treatment. As a result, it is needed to improve drug bioavailability in the tumor region while being restricted to the appointed target and cause a reduction in the amount of prescribed drug, which decreases the number and severity of side effects and leads to the delivery of proper and sufficient dosage of combined drugs to the designated target through a controlled manner. Due to a variety of unique physiochemical properties, numerous studies were performed on Au nanoparticles (Au NPs) for the objectives of drug delivery, radiosensitizers, and photothermal and photodynamic therapy agents. It is also notable that the controllable production of nanomaterials can provide products with controlled sizes and shapes. There are a variety of clinical applications for the unique optical, chemical, and biological features of  Au nanoparticles that involve drug and gene delivery. Among the other appealing qualities of these nanoparticles, one can point out the intensification of their surface plasmon, the ease of surface modification, accommodation of a controlled method that provides their interaction with thiol groups, and their non-toxic nature. These properties can be utilized to develop a platform for achieving an effective and selective design for performing the targeted intracellular release of particular drugs [128-130]. The recent progresses of Au nanoparticles exertion in drug and Therapeutic agents delivery systems is presented in Table 3.

Theranostics based on Au nanoparticles in ovarian cancer
Nanotheranostics refer to the combination of optical multiplexed disease detection and therapeutic monitoring in a single modality, with the ability to escalate nanomedicine closer to a true form of personalized medicine [152]. The delivery of efficient treatments to cancer patients is beginning to face alterations due to the development of targeted cancer therapies, which implicate the rise of nanomedicine as a cutting-edge field in regards to the objective of personalized medicine[74]. The amazing potential of AuNPs in the roles of drug delivery, diagnostics, and imaging carriers is undeniable. Multifunctional hybrid AuNPs were designed for multimodal imaging, diagnosis, and treatment optimization. AuNPs are divided into several subtypes and despite their variances, some of their distinct types can be an excellent choice for specific cancer theranostics applications due to their different features. Next to being biocompatible, AuNPS contain a strong surface plasmon resonance (SPR) peak with the option of being tuned from visible to near-infrared (NIR), which can be easily functionalized through the addition of various molecules to their surfaces [153, 154]. The accommodation of a diverse and straightforward chemistry facilities the simple attaching of numerous biomolecules to the surfaces of these nanoparticles and therefore, they can be a suitable option for cancer theranostics. Peptides, small-molecular-weight chemicals, aptamers, and monoclonal antibodies were added to the surface of AuNPs to improve their tumor-targeting capabilities. The interaction of an electromagnetic wave with conduction electrons results in the creation of a metal, which is an optical phenomenon known as SPR and stands as another attribute of AuNPs. This product has been exerted for multimodality imaging, tumor targeting, and therapeutic delivery(Fig. 1). Au nanoparticles can also function as anticancer drug carriers due to their enhanced permeability and retention (EPR) effects that facilitate their aggregation at tumor sites. Furthermore, the surface of Au nanoparticles can be easily modified to provide the binding of targeted groups and offer active targeting properties to the carriers. Moreover, the rate of drug bioavailability and treatment efficacy can be improved by the application of Au nanoparticles as carriers. Drugs and genes can be simultaneously delivered through the usage of functionalized Au nanoparticle[162-164]. Also, delivery systems based on Au nanoparticles can intelligently release drugs and gene by considering the redox sensitivity, pH sensitivity, thermo-sensitivity, and other factors [165]. Table 4 demonstrates some of the theranostic applications based on Au nanoparticles in ovarian cancer.

THERANOSTIC  & FUTURE PERSPECTIVES
There are unquestionable proofs for the great potential of AuNPs in functioning as drug delivery, diagnostics, and imaging vectors. However, their rate of progress is still considerably slower than that of desired and despite the significant attempts for introducing AuNPs into clinical frameworks, only a few clinical trials have been conducted. Although this product is typically regarded to be biocompatible, yet a number of studies reported the observance of toxicity based on size, shape, and surface coating among the varying parameters. Other concerns implicate their limited penetration depth that inhibits imaging and PTT implementation, non-biodegradability that can result in bioaccumulation in various tissues and organs, and non-porousness that might cause bioaccumulation in a variety of tissues and organs and turn drug encapsulation and release into a challenge [166, 167]. Nevertheless, comprehensive characterization is required since the pharmacokinetics and biocompatibility of AuNPs can be different depending on the type of applied conjugation. The conduction of further long-term toxicity studies, as well as detailed assessments into the mechanisms of AuNPs interaction with biological systems. stand as a necessity and must be increased. Similar to nano theranostics techniques and their application in clinics, there is still a long way before the complete preparation and availability of nanotechnology platforms for the usage of clinicians[74, 168].

CONCLUSIONS
Au nanoparticles proved to contain a high potential for diagnosis, treating and improving the living conditions of patients with ovarian cancer. Gold nanoparticles can be exerted in a variety of methods for the early detection of this disease. They can be also integrated with different imaging modes and provide a more accurate diagnosis of  cancer, which is a crucial factor in the success of following treatments. Using novel methods of cancer treatment instead of traditional procedures (surgery - chemotherapy) can lead to a reduction in the unwanted side effects of chemotherapy, surpassing the obstacle of drug resistance, and preventing disease recurrence to some extent in order to avert the death of patients. It is expected that a faster and greater rate of improvement would be achieved through the utilization of Au nanoparticles as carriers for drug delivery and other therapeutic agents throughout the treatment of patients with ovarian cancer. In fact, this product can prevent the inducement of unwanted toxicity to normal tissues by transporting the drug and its delivery to the desired location, which would consequently cause more toxicity to the target site. Furthermore, the application of Au nanoparticles can guarantee a longer circulation in the blood, which would improve the obtained therapeutic efficiency when combined with successful targeting. Gold nanoparticles can stand as a new horizon for the integration of diagnostic and treatment methods and facilitate the design of personalized medicine, which emphasizes on the necessity of further comprehensive research to demonstrate its effectiveness.

ACKNOWLEDGEMENT
The figure was designed on the BioRender website. The authors thank the designers of this website.

CONFLICT OF INTEREST
The authors declare no conflicts of interest.

1. Acmaz G, Aksoy H, Albayrak E, Baser M, Ozyurt S, Aksoy U, et al. Evaluation of Endometrial Precancerous Lesions in Postmenopausal Obese Women - A High Risk Group? Asian Pacific Journal of Cancer Prevention. 2014;15(1):195-8.
2. Eckert MA, Pan S, Hernandez KM, Loth RM, Andrade J, Volchenboum SL, et al. Genomics of Ovarian Cancer Progression Reveals Diverse Metastatic Trajectories Including Intraepithelial Metastasis to the Fallopian Tube. Cancer Discov. 2016;6(12):1342-51.
3.    Fathalla, M., Incessant ovulation and ovarian cancer–a hypothesis re-visited. Facts, views & vision in ObGyn, 2013. 5(4): p. 292.
4.    Matz, M., et al., Worldwide comparison of ovarian cancer survival: Histological group and stage at diagnosis (CONCORD-2). Gynecologic oncology, 2017. 144(2): p. 396-404.
5. Zhao L, Li Y, Zhang Z, Zou J, Li J, Wei R, et al. Meta-analysis based gene expression profiling reveals functional genes in ovarian cancer. Biosci Rep. 2020;40(11):BSR20202911.
6. Dumas L, Lidington E, Appadu L, Jupp P, Husson O, Banerjee S. Exploring Older Women’s Attitudes to and Experience of Treatment for Advanced Ovarian Cancer: A Qualitative Phenomenological Study. Cancers (Basel). 2021;13(6):1207.
7. Alcaraz-Sanabria A, Baliu-Piqué M, Saiz-Ladera C, Rojas K, Manzano A, Marquina G, et al. Genomic Signatures of Immune Activation Predict Outcome in Advanced Stages of Ovarian Cancer and Basal-Like Breast Tumors. Front Oncol. 2020;9:1486-.
8.    Reid, B.M., J.B. Permuth, and T.A. Sellers, Epidemiology of ovarian cancer: a review. Cancer biology & medicine, 2017. 14(1): p. 9.
9. Moore KN, Secord AA, Geller MA, Miller DS, Cloven N, Fleming GF, et al. Niraparib monotherapy for late-line treatment of ovarian cancer (QUADRA): a multicentre, open-label, single-arm, phase 2 trial. The Lancet Oncology. 2019;20(5):636-48.
10. Barani M, Bilal M, Sabir F, Rahdar A, Kyzas GZ. Nanotechnology in ovarian cancer: Diagnosis and treatment. Life Sciences. 2021;266:118914.
11.    Mekradh, H.A. and F.M. Hameed, Gold Nanoparticles as anti-ovarian cancer therapy. Al-Qadisiyah Medical Journal, 2019. 15(2).
12.    Henderson, J.T., E.M. Webber, and G.F. Sawaya, Screening for ovarian cancer: updated evidence report and systematic review for the US preventive services task force. Jama, 2018. 319(6): p. 595-606.
13. Tomasińska A, Stukan M, Badocha M, Myszewska A. Accuracy of Pretreatment Ultrasonography Assessment of Intra-Abdominal Spread in Epithelial Ovarian Cancer: A Prospective Study. Diagnostics (Basel). 2021;11(9):1600.
14. Jelovac D, Armstrong DK. Recent progress in the diagnosis and treatment of ovarian cancer. CA: a cancer journal for clinicians. 2011;61(3):183-203.
15. Mansha M, Gill A, Thomson PC. Potential risk factors of ovarian cancer and analysis of CA125, a biomarker used for its monitoring and diagnosis. Molecular Biology Reports. 2019;46(3):3325-32.
16. McMullen M, Karakasis K, Rottapel R, Oza AM. Advances in ovarian cancer, from biology to treatment. Nature Cancer. 2021;2(1):6-8.
17. Cortez AJ, Tudrej P, Kujawa KA, Lisowska KM. Advances in ovarian cancer therapy. Cancer chemotherapy and pharmacology. 2018;81(1):17-38.
18. Lheureux S, Gourley C, Vergote I, Oza AM. Epithelial ovarian cancer. The Lancet. 2019;393(10177):1240-53.
19. Mutch DG, Prat J. 2014 FIGO staging for ovarian, fallopian tube and peritoneal cancer. Gynecologic Oncology. 2014;133(3):401-4.
20. Webb PM, Jordan SJ. Epidemiology of epithelial ovarian cancer. Best Practice & Research Clinical Obstetrics & Gynaecology. 2017;41:3-14.
21. Chen S-N, Chang R, Lin L-T, Chern C-U, Tsai H-W, Wen Z-H, et al. MicroRNA in Ovarian Cancer: Biology, Pathogenesis, and Therapeutic Opportunities. Int J Environ Res Public Health. 2019;16(9):1510.
22. Matulonis UA, Sood AK, Fallowfield L, Howitt BE, Sehouli J, Karlan BY. Ovarian cancer. Nat Rev Dis Primers. 2016;2:16061-.
23. Peres LC, Cushing-Haugen KL, Köbel M, Harris HR, Berchuck A, Rossing MA, et al. Invasive Epithelial Ovarian Cancer Survival by Histotype and Disease Stage. J Natl Cancer Inst. 2019;111(1):60-8.
24. Bryant A, Hiu S, Kunonga P, Gajjar K, Craig D, Vale L, et al. Impact of residual disease as a prognostic factor for survival in women with advanced epithelial ovarian cancer after primary surgery. Cochrane Database of Systematic Reviews. 2021;2021(9).
25. Mueller JJ, Zhou QC, Iasonos A, O’Cearbhaill RE, Alvi FA, El Haraki A, et al. Neoadjuvant chemotherapy and primary debulking surgery utilization for advanced-stage ovarian cancer at a comprehensive cancer center. Gynecologic oncology. 2016;140(3):436-42.
26. Dai J, Cheng Y, Wu J, Wang Q, Wang W, Yang J, et al. Modular Peptide Probe for Pre/Intra/Postoperative Therapeutic to Reduce Recurrence in Ovarian Cancer. ACS Nano. 2020;14(11):14698-714.
27. Hernandez-Lopez LA, Elizalde-Mendez A. How far should we go in optimal cytoreductive surgery for ovarian cancer? Chinese Clinical Oncology. 2020;9(5):70-.
28. Bartlett DL, Howe JR, Chang G, Crago A, Hogg M, Karakousis G, et al. Management of Cancer Surgery Cases During the COVID-19 Pandemic: Considerations. Ann Surg Oncol. 2020;27(6):1717-20.
29. Jiang R, Zhu J, Kim JW, Liu J, Kato K, Kim HS, et al. Study of upfront surgery versus neoadjuvant chemotherapy followed by interval debulking surgery for patients with stage IIIC and IV ovarian cancer, SGOG SUNNY (SOC-2) trial concept. J Gynecol Oncol. 2020;31(5):e86-e.
30. Trimbos JB. Surgical treatment of early-stage ovarian cancer. Best Practice & Research Clinical Obstetrics & Gynaecology. 2017;41:60-70.
31. Vitale SG, Marilli I, Lodato M, Tropea A, Cianci A. The role of cytoreductive surgery in advanced-stage ovarian cancer: a systematic review. Updates in Surgery. 2013;65(4):265-70.
32. Lheureux S, Braunstein M, Oza AM. Epithelial ovarian cancer: Evolution of management in the era of precision medicine. CA: A Cancer Journal for Clinicians. 2019.
33. Narod S. Can advanced-stage ovarian cancer be cured? Nature Reviews Clinical Oncology. 2016;13(4):255-61.
34. Nick AM, Coleman RL, Ramirez PT, Sood AK. A framework for a personalized surgical approach to ovarian cancer. Nat Rev Clin Oncol. 2015;12(4):239-45.
35. Sánchez-Hidalgo JM, Rodríguez-Ortiz L, Arjona-Sánchez Á, Rufián-Peña S, Casado-Adam Á, Cosano-Álvarez A, et al. Colorectal peritoneal metastases: Optimal management review. World J Gastroenterol. 2019;25(27):3484-502.
36. Pokhriyal R, Hariprasad R, Kumar L, Hariprasad G. Chemotherapy Resistance in Advanced Ovarian Cancer Patients. Biomark Cancer. 2019;11:1179299X19860815-1179299X.
37. Mirnezami SMS, Heydarinasab A, Akbarzadeh A, Arjmand M. Investigation of characterization and cytotoxic effect of PEGylated nanoliposomal containing Melphalan on ovarian cancer: an in vitro study. Journal of Experimental Nanoscience. 2021;16(1):101-15.
38. Wang Q, Ren X, Liu P, Li J, Lv J, Wang J, et al. Improved genome assembly of Chinese shrimp ( Fenneropenaeus chinensis ) suggests adaptation to the environment during evolution and domestication. Molecular Ecology Resources. 2021;22(1):334-44.
39. Gore M, Hackshaw A, Brady WE, Penson RT, Zaino R, McCluggage WG, et al. An international, phase III randomized trial in patients with mucinous epithelial ovarian cancer (mEOC/GOG 0241) with long-term follow-up: and experience of conducting a clinical trial in a rare gynecological tumor. Gynecologic oncology. 2019;153(3):541-8.
40. Podwika SE, Jenkins TM, Khokhar JK, Erickson SH, Modesitt SC. Optimal age for genetic cancer predisposition testing in hereditary SMARCA4 Ovarian Cancer Families: How young is too young? Gynecol Oncol Rep. 2020;32:100569-.
41. Cheng M, Lee HH, Chang W-H, Lee N-R, Huang H-Y, Chen Y-J, et al. Weekly Dose-Dense Paclitaxel and Triweekly Low-Dose Cisplatin: A Well-Tolerated and Effective Chemotherapeutic Regimen for First-Line Treatment of Advanced Ovarian, Fallopian Tube, and Primary Peritoneal Cancer. Int J Environ Res Public Health. 2019;16(23):4794.
42. Huang C-Y, Cheng M, Lee N-R, Huang H-Y, Lee W-L, Chang W-H, et al. Comparing Paclitaxel-Carboplatin with Paclitaxel-Cisplatin as the Front-Line Chemotherapy for Patients with FIGO IIIC Serous-Type Tubo-Ovarian Cancer. Int J Environ Res Public Health. 2020;17(7):2213.
43. Hakeam H, Ayman A, Waleed AT, Amen T. Systemic complications of the bidirectional intraoperative chemotherapy with intravenous ifosfamide and hyperthermic intraperitoneal chemotherapy (HIPEC) using cisplatin plus doxorubicin. Pleura Peritoneum. 2019;4(4):20190025-.
44. Wu R, Zhang Z, Wang B, Chen G, Zhang Y, Deng H, et al. Combination Chemotherapy of Lung Cancer - Co-Delivery of Docetaxel Prodrug and Cisplatin Using Aptamer-Decorated Lipid-Polymer Hybrid Nanoparticles. Drug Des Devel Ther. 2020;14:2249-61.
45. Schneeweiss A, Michel LL, Möbus V, Tesch H, Klare P, Hahnen E, et al. Survival analysis of the randomised phase III GeparOcto trial comparing neoadjuvant chemotherapy of intense dose-dense epirubicin, paclitaxel, cyclophosphamide versus weekly paclitaxel, liposomal doxorubicin (plus carboplatin in triple-negative breast cancer) for patients with high-risk early breast cancer. European Journal of Cancer. 2022;160:100-11.
46. Kim J-H, Yoon YS, Kim JC, Kim Y-M. Assessment of the Applicability of Integrative Tumor Response Assays in Advanced Epithelial Ovarian Cancer. Anticancer Research. 2018;39(1):313-8.
47. Lindemann K, Beale PJ, Rossi E, Goh JC, Vaughan MM, Tenney ME, et al. Phase I study of BNC105P, carboplatin and gemcitabine in partially platinum-sensitive ovarian cancer patients in first or second relapse (ANZGOG-1103). Cancer Chemotherapy and Pharmacology. 2018;83(1):97-105.
48. Walsh CS, Kamrava M, Rogatko A, Kim S, Li A, Cass I, et al. Phase II trial of cisplatin, gemcitabine and pembrolizumab for platinum-resistant ovarian cancer. PLoS One. 2021;16(6):e0252665-e.
49. Cohen S, Schwartz M, Dottino P, Beddoe AM. Use of a multi-drug regimen gemcitabine, 5-fluorouracil, irinotecan, cisplatin, bevacizumab, docetaxel, and cyclophosphamide (GFIP/BDC) for heavily pretreated relapsed epithelial ovarian, fallopian tube and primary peritoneal cancer. J Ovarian Res. 2019;12(1):36-.
50. Weng C-S, Wu C-C, Chen T-C, Chen J-R, Huang C-Y, Chang C-L. Retrospective Analysis Of Comparative Outcomes In Recurrent Platinum-Sensitive Ovarian Cancer Treated With Pegylated Liposomal Doxorubicin (Lipo-Dox) And Carboplatin Versus Paclitaxel And Carboplatin. Cancer Manag Res. 2019;11:9899-905.
51. Lee M-W, Ryu H, Song I-C, Yun H-J, Jo D-Y, Ko YB, et al. Efficacy of cisplatin combined with topotecan in patients with advanced or recurrent ovarian cancer as second- or higher-line palliative chemotherapy. Medicine (Baltimore). 2020;99(17):e19931-e.
52.    Harter, P., et al., Randomized Trial of Cytoreductive Surgery for Relapsed Ovarian Cancer. New England Journal of Medicine, 2021. 385(23): p. 2123-2131.
53. Jaaback K, Johnson N, Lawrie TA. Intraperitoneal chemotherapy for the initial management of primary epithelial ovarian cancer. Cochrane Database Syst Rev. 2011(11):CD005340-CD.
54. Yallapu MM, Jaggi M, Chauhan SC. Scope of nanotechnology in ovarian cancer therapeutics. J Ovarian Res. 2010;3:19-.
55. Liu Y-Q, Wang X-L, He D-H, Cheng Y-X. Protection against chemotherapy- and radiotherapy-induced side effects: A review based on the mechanisms and therapeutic opportunities of phytochemicals. Phytomedicine. 2021;80:153402.
56. Yap TA, Carden CP, Kaye SB. Beyond chemotherapy: targeted therapies in ovarian cancer. Nature Reviews Cancer. 2009;9(3):167-81.
57. Majidinia M, Alizadeh E, Yousefi B, Akbarzadeh M, Zarghami N. Downregulation of Notch Signaling Pathway as an Effective Chemosensitizer for Cancer Treatment. Drug Research. 2016;66(11):571-9.
58. Bukowski K, Kciuk M, Kontek R. Mechanisms of Multidrug Resistance in Cancer Chemotherapy. Int J Mol Sci. 2020;21(9):3233.
59. Tabrizi AE, Tahmoorespur M, Ebrahimie E. ABC Transporters are Hub Genes in Response of Resistant E. Coli ST131 to Ciprofloxacin. Archives Of Pharmacy Practice. 2021;12(3):82-8.
60. Anand SK, Sharma A, Singh N, Kakkar P. Entrenching role of cell cycle checkpoints and autophagy for maintenance of genomic integrity. DNA Repair. 2020;86:102748.
61. Zeng Y, Ma J, Xu L, Wu D. Natural Product Gossypol and its Derivatives in Precision Cancer Medicine. Current Medicinal Chemistry. 2019;26(10):1849-73.
62. Jiang W, Xia J, Xie S, Zou R, Pan S, Wang Z-w, et al. Long non-coding RNAs as a determinant of cancer drug resistance: Towards the overcoming of chemoresistance via modulation of lncRNAs. Drug Resistance Updates. 2020;50:100683.
63. Moioli M, Barra F, Maramai M, Valenzano Menada M, Vellone VG, Costantini S, et al. Mucinous ovarian cancer: current therapeutic targets, preclinical progress, and experimental drugs. Expert Opinion on Investigational Drugs. 2019;28(12):1025-9.
64. Gadducci A, Guarneri V, Peccatori FA, Ronzino G, Scandurra G, Zamagni C, et al. Current strategies for the targeted treatment of high-grade serous epithelial ovarian cancer and relevance of BRCA mutational status. J Ovarian Res. 2019;12(1):9-.
65. Duan Y-T, Sangani CB, Liu W, Soni KV, Yao Y. New Promises to Cure Cancer and Other Genetic Diseases/Disorders: Epi-drugs Through Epigenetics. Current Topics in Medicinal Chemistry. 2019;19(12):972-94.
66. Hainsworth JD, Kalman L, Castine M, Sylvester L, Anthony Greco F. Paclitaxel, carboplatin, and oral etoposide as initial treatment for advanced ovarian carcinoma: a Minnie Pearl Cancer Research Network phase II trial. Gynecologic Oncology. 2005;97(1):200-5.
67. Satpathy M, Wang L, Zielinski RJ, Qian W, Wang YA, Mohs AM, et al. Targeted Drug Delivery and Image-Guided Therapy of Heterogeneous Ovarian Cancer Using HER2-Targeted Theranostic Nanoparticles. Theranostics. 2019;9(3):778-95.
68. Jhawat V, Gulia M, Gupta S, Maddiboyina B, Dutt R. Integration of pharmacogenomics and theranostics with nanotechnology as quality by design (QbD) approach for formulation development of novel dosage forms for effective drug therapy. Journal of Controlled Release. 2020;327:500-11.
69. Seca C, Ferraresi A, Phadngam S, Vidoni C, Isidoro C. Autophagy-dependent toxicity of amino-functionalized nanoparticles in ovarian cancer cells. Journal of Materials Chemistry B. 2019;7(35):5376-91.
70. Talluri S, Malla RR. Superparamagnetic Iron Oxide Nanoparticles (SPIONs) for Diagnosis and Treatment of Breast, Ovarian and Cervical Cancers. Current Drug Metabolism. 2020;20(12):942-5.
71. Navya PN, Kaphle A, Srinivas SP, Bhargava SK, Rotello VM, Daima HK. Current trends and challenges in cancer management and therapy using designer nanomaterials. Nano Converg. 2019;6(1):23-.
72. Santos AC, Pereira I, Pereira-Silva M, Ferreira L, Caldas M, Magalhães M, et al. Nanocarriers for resveratrol delivery: Impact on stability and solubility concerns. Trends in Food Science & Technology. 2019;91:483-97.
73. Ashfaq UA, Riaz M, Yasmeen E, Yousaf MZ. Recent Advances in Nanoparticle-Based Targeted Drug-Delivery Systems Against Cancer and Role of Tumor Microenvironment. Critical Reviews in Therapeutic Drug Carrier Systems. 2017;34(4):317-53.
74. Cabral RM, Baptista PV. Anti-cancer precision theranostics: a focus on multifunctional gold nanoparticles. Expert Review of Molecular Diagnostics. 2014;14(8):1041-52.
75. Arvizo RR, Saha S, Wang E, Robertson JD, Bhattacharya R, Mukherjee P. Inhibition of tumor growth and metastasis by a self-therapeutic nanoparticle. Proc Natl Acad Sci U S A. 2013;110(17):6700-5.
76. Chauhan VP, Stylianopoulos T, Martin JD, Popović Z, Chen O, Kamoun WS, et al. Normalization of tumour blood vessels improves the delivery of nanomedicines in a size-dependent manner. Nat Nanotechnol. 2012;7(6):383-8.
77. Lee KX, Shameli K, Yew YP, Teow S-Y, Jahangirian H, Rafiee-Moghaddam R, et al. Recent Developments in the Facile Bio-Synthesis of Gold Nanoparticles (AuNPs) and Their Biomedical Applications. Int J Nanomedicine. 2020;15:275-300.
78. Lee KD, Nagajyothi PC, Sreekanth TVM, Park S. Eco-friendly synthesis of gold nanoparticles (AuNPs) using Inonotus obliquus and their antibacterial, antioxidant and cytotoxic activities. Journal of Industrial and Engineering Chemistry. 2015;26:67-72.
79. Esfahani AR, Sadiq Z, Oyewunmi OD, Safiabadi Tali SH, Usen N, Boffito DC, et al. Portable, stable, and sensitive assay to detect phosphate in water with gold nanoparticles (AuNPs) and dextran tablet. The Analyst. 2021;146(11):3697-708.
80. Dong H, Zou F, Hu X, Zhu H, Koh K, Chen H. Analyte induced AuNPs aggregation enhanced surface plasmon resonance for sensitive detection of paraquat. Biosensors and Bioelectronics. 2018;117:605-12.
81. Ibrahim NA, Eid BM, Abdel-Aziz MS. Green synthesis of AuNPs for eco-friendly functionalization of cellulosic substrates. Applied Surface Science. 2016;389:118-25.
82. Mbambo MC, Madito MJ, Khamliche T, Mtshali CB, Khumalo ZM, Madiba IG, et al. Thermal conductivity enhancement in gold decorated graphene nanosheets in ethylene glycol based nanofluid. Sci Rep. 2020;10(1):14730-.
83. Rizwan M, Elma S, Lim SA, Ahmed MU. AuNPs/CNOs/SWCNTs/chitosan-nanocomposite modified electrochemical sensor for the label-free detection of carcinoembryonic antigen. Biosensors and Bioelectronics. 2018;107:211-7.
84. Ahmed HB, El-Hawary NS, Emam HE. Self-assembled AuNPs for ingrain pigmentation of silk fabrics with antibacterial potency. International Journal of Biological Macromolecules. 2017;105:720-9.
85. Li W, Cao Z, Liu R, Liu L, Li H, Li X, et al. AuNPs as an important inorganic nanoparticle applied in drug carrier systems. Artificial Cells, Nanomedicine, and Biotechnology. 2019;47(1):4222-33.
86. Amdouni S, Cherifi Y, Coffinier Y, Addad A, Zaïbi MA, Oueslati M, et al. Gold nanoparticles coated silicon nanowires for efficient catalytic and photocatalytic applications. Materials Science in Semiconductor Processing. 2018;75:206-13.
87. Su Y, Wei X, Peng F, Zhong Y, Lu Y, Su S, et al. Gold Nanoparticles-Decorated Silicon Nanowires as Highly Efficient Near-Infrared Hyperthermia Agents for Cancer Cells Destruction. Nano Letters. 2012;12(4):1845-50.
88. Zhao Z, Sun Y, Li P, Zhang W, Lian K, Hu J, et al. Preparation and characterization of AuNPs/CNTs-ErGO electrochemical sensors for highly sensitive detection of hydrazine. Talanta. 2016;158:283-91.
89. Nair KG, Vishnuraj R, Pullithadathil B. Integrated co-axial electrospinning for a single-step production of 1D aligned bimetallic carbon fibers@AuNPs–PtNPs/NiNPs–PtNPs towards H2 detection. Materials Advances. 2022;3(1):443-55.
90. Saleh TA, Al-Shalalfeh MM, Onawole AT, Al-Saadi AA. Ultra-trace detection of methimazole by surface-enhanced Raman spectroscopy using gold substrate. Vibrational Spectroscopy. 2017;90:96-103.
91. V R, Pal K, Zaheer T, Kalarikkal N, Thomas S, de Souza FG, et al. Gold nanoparticles against respiratory diseases: oncogenic and viral pathogens review. Therapeutic Delivery. 2020;11(8):521-34.
92. Abdelhamid HN, Wu H-F. Gold nanoparticles assisted laser desorption/ionization mass spectrometry and applications: from simple molecules to intact cells. Analytical and Bioanalytical Chemistry. 2016;408(17):4485-502.
93. Baptista PV. Gold nanobeacons: a potential nanotheranostics platform. Nanomedicine. 2014;9(15):2247-50.
94. Borse VB, Konwar AN, Jayant RD, Patil PO. Perspectives of characterization and bioconjugation of gold nanoparticles and their application in lateral flow immunosensing. Drug Delivery and Translational Research. 2020;10(4):878-902.
95. Chen H, Liu Z, Li S, Su C, Qiu X, Zhong H, et al. Fabrication of Graphene and AuNP Core Polyaniline Shell Nanocomposites as Multifunctional Theranostic Platforms for SERS Real-time Monitoring and Chemo-photothermal Therapy. Theranostics. 2016;6(8):1096-104.
96. Bai X, Wang Y, Song Z, Feng Y, Chen Y, Zhang D, et al. The Basic Properties of Gold Nanoparticles and their Applications in Tumor Diagnosis and Treatment. Int J Mol Sci. 2020;21(7):2480.
97. Chatterjee M, Hurley LC, Tainsky MA. Paraneoplastic antigens as biomarkers for early diagnosis of ovarian cancer. Gynecol Oncol Rep. 2017;21:37-44.
98. Attia MF, Wallyn J, Anton N, Vandamme TF. Inorganic Nanoparticles for X-Ray Computed Tomography Imaging. Critical Reviews™ in Therapeutic Drug Carrier Systems. 2018;35(5):391-431.
99. Suarasan S, Licarete E, Astilean S, Craciun A-M. Probing cellular uptake and tracking of differently shaped gelatin-coated gold nanoparticles inside of ovarian cancer cells by two-photon excited photoluminescence analyzed by fluorescence lifetime imaging (FLIM). Colloids and Surfaces B: Biointerfaces. 2018;166:135-43.
100. Wu Y, Ali MRK, Chen K, Fang N, El-Sayed MA. Gold nanoparticles in biological optical imaging. Nano Today. 2019;24:120-40.
101. Shen R, Zhang J, Huang W, Wu S, Li G, Zou S, et al. Dynamic light scattering and fluorescence dual-signal sensing of cancer antigen-125 via recognition of the polymerase chain reaction product with gold nanoparticle probe. Analytica Chimica Acta. 2021;1145:87-94.
102. Hada A-M, Potara M, Suarasan S, Vulpoi A, Nagy-Simon T, Licarete E, et al. Fabrication of gold–silver core–shell nanoparticles for performing as ultrabright SERS-nanotags inside human ovarian cancer cells. Nanotechnology. 2019;30(31):315701.
103. TunÇ İ, Susapto HH. Label-Free Detection of Ovarian Cancer Antigen CA125 by Surface Enhanced Raman Scattering. Journal of Nanoscience and Nanotechnology. 2020;20(3):1358-65.
104. Liu X, Wang Q, Chen J, Chen X, Yang W. Ultrasensitive electrochemiluminescence biosensor for the detection of tumor exosomes based on peptide recognition and luminol-AuNPs@g-C3N4 nanoprobe signal amplification. Talanta. 2021;221:121379.
105. Zhang P, Wang J, Huang H, Yu B, Qiu K, Huang J, et al. Unexpected high photothemal conversion efficiency of gold nanospheres upon grafting with two-photon luminescent ruthenium(II) complexes: A way towards cancer therapy? Biomaterials. 2015;63:102-14.
106. Wang Z, Sun X, Huang T, Song J, Wang Y. A Sandwich Nanostructure of Gold Nanoparticle Coated Reduced Graphene Oxide for Photoacoustic Imaging-Guided Photothermal Therapy in the Second NIR Window. Front Bioeng Biotechnol. 2020;8:655-.
107. Manivasagan P, Bharathiraja S, Bui NQ, Lim IG, Oh J. Paclitaxel-loaded chitosan oligosaccharide-stabilized gold nanoparticles as novel agents for drug delivery and photoacoustic imaging of cancer cells. International Journal of Pharmaceutics. 2016;511(1):367-79.
108. Dongying Q, Lan L, Qian D. Targeting of ovarian cancer cell through functionalized gold nanoparticles by novel glypican-3- binding peptide as a ultrasound contrast agents. Process Biochemistry. 2020;98:51-8.
109. Ghaziyani MF, Pourhassan Moghaddam M, Shahbazi-Gahrouei D, Ghavami M, Mohammadi A, Abbasi MM, et al. Anti-CD24 bio Modified PEGylated Gold Nanoparticles as Targeted Computed Tomography Contrast Agent. Adv Pharm Bull. 2018;8(4):599-607.
110. Lin J, Hu W, Gao F, Qin J, Peng C, Lu X. Folic acid-modified diatrizoic acid-linked dendrimer-entrapped gold nanoparticles enable targeted CT imaging of human cervical cancer. J Cancer. 2018;9(3):564-77.
111. Gong T, Kong KV, Goh D, Olivo M, Yong K-T. Sensitive surface enhanced Raman scattering multiplexed detection of matrix metalloproteinase 2 and 7 cancer markers. Biomed Opt Express. 2015;6(6):2076-87.
112. Joseph MM, Nair JB, Maiti KK, Therakathinal T S. Plasmonically Enhanced Galactoxyloglucan Endowed Gold Nanoparticles Exposed Tumor Targeting Biodistribution Envisaged in a Surface-Enhanced Raman Scattering Platform. Biomacromolecules. 2017;18(12):4041-53.
113. Li S, Hu C, Chen C, Zhang J, Bai Y, Tan CS, et al. Molybdenum Disulfide Supported on Metal–Organic Frameworks as an Ultrasensitive Layer for the Electrochemical Detection of the Ovarian Cancer Biomarker CA125. ACS Applied Bio Materials. 2021;4(7):5494-502.
114. Crulhas BP, Basso CR, Castro GR, Pedrosa VA. Review—Recent Advances Based on a Sensor for Cancer Biomarker Detection. ECS Journal of Solid State Science and Technology. 2021;10(4):047004.
115. Feng D, Su J, He G, Xu Y, Wang C, Zheng M, et al. Electrochemical DNA Sensor for Sensitive BRCA1 Detection Based on DNA Tetrahedral-Structured Probe and Poly-Adenine Mediated Gold Nanoparticles. Biosensors (Basel). 2020;10(7):78.
116. Elshafey R, Tavares AC, Siaj M, Zourob M. Electrochemical impedance immunosensor based on gold nanoparticles–protein G for the detection of cancer marker epidermal growth factor receptor in human plasma and brain tissue. Biosensors and Bioelectronics. 2013;50:143-9.
117. Hasanzadeh M, Sahmani R, Solhi E, Mokhtarzadeh A, Shadjou N, Mahboob S. Ultrasensitive immunoassay of carcinoma antigen 125 in untreated human plasma samples using gold nanoparticles with flower like morphology: A new platform in early stage diagnosis of ovarian cancer and efficient management. International Journal of Biological Macromolecules. 2018;119:913-25.
118. Gidwani B, Vyas A. The potentials of nanotechnology-based drug delivery system for treatment of ovarian cancer. Artificial Cells, Nanomedicine, and Biotechnology. 2013;43(4):291-7.
119. Shah K, Rawal RM. Genetic and Epigenetic Modulation of Drug Resistance in Cancer: Challenges and Opportunities. Current Drug Metabolism. 2020;20(14):1114-31.
120. Zhang Z, Zhou L, Xie N, Nice EC, Zhang T, Cui Y, et al. Overcoming cancer therapeutic bottleneck by drug repurposing. Signal Transduct Target Ther. 2020;5(1):113-.
121. Khan AU, Khan M, Cho MH, Khan MM. Selected nanotechnologies and nanostructures for drug delivery, nanomedicine and cure. Bioprocess and Biosystems Engineering. 2020;43(8):1339-57.
122. Terraneo N, Jacob F, Dubrovska A, Grünberg J. Novel Therapeutic Strategies for Ovarian Cancer Stem Cells. Front Oncol. 2020;10:319-.
123. Bahrami B, Hojjat-Farsangi M, Mohammadi H, Anvari E, Ghalamfarsa G, Yousefi M, et al. Nanoparticles and targeted drug delivery in cancer therapy. Immunology Letters. 2017;190:64-83.
124. Shariatinia Z. Big family of nano- and microscale drug delivery systems ranging from inorganic materials to polymeric and stimuli-responsive carriers as well as drug-conjugates. Journal of Drug Delivery Science and Technology. 2021;66:102790.
125. Haider M, Abdin SM, Kamal L, Orive G. Nanostructured Lipid Carriers for Delivery of Chemotherapeutics: A Review. Pharmaceutics. 2020;12(3):288.
126. Amina SJ, Guo B. A Review on the Synthesis and Functionalization of Gold Nanoparticles as a Drug Delivery Vehicle. Int J Nanomedicine. 2020;15:9823-57.
127. Mahmoud BS, AlAmri AH, McConville C. Polymeric Nanoparticles for the Treatment of Malignant Gliomas. Cancers (Basel). 2020;12(1):175.
128. Piktel E, Ościłowska I, Suprewicz Ł, Depciuch J, Marcińczyk N, Chabielska E, et al. ROS-Mediated Apoptosis and Autophagy in Ovarian Cancer Cells Treated with Peanut-Shaped Gold Nanoparticles. Int J Nanomedicine. 2021;16:1993-2011.
129. Yang W, Liang H, Ma S, Wang D, Huang J. Gold nanoparticle based photothermal therapy: Development and application for effective cancer treatment. Sustainable Materials and Technologies. 2019;22:e00109.
130. Yao Y, Zhou Y, Liu L, Xu Y, Chen Q, Wang Y, et al. Nanoparticle-Based Drug Delivery in Cancer Therapy and Its Role in Overcoming Drug Resistance. Front Mol Biosci. 2020;7:193-.
131. Xiong X, Arvizo RR, Saha S, Robertson DJ, McMeekin S, Bhattacharya R, et al. Sensitization of ovarian cancer cells to cisplatin by gold nanoparticles. Oncotarget. 2014;5(15):6453-65.
132. Rezaee Z, Yadollahpour A, Bayati V, Negad Dehbashi F. Gold nanoparticles and electroporation impose both separate and synergistic radiosensitizing effects in HT-29 tumor cells: an in vitro study. Int J Nanomedicine. 2017;12:1431-9.
133. Akbal Vural O, Yaman YT, Bolat G, Abaci S. Human Serum Albumin−Gold Nanoparticle Based Impedimetric Sensor for Sensitive Detection of miRNA‐200c. Electroanalysis. 2020;33(4):925-35.
134. Kip B, Tunc CU, Aydin O. Triple-combination therapy assisted with ultrasound-active gold nanoparticles and ultrasound therapy against 3D cisplatin-resistant ovarian cancer model. Ultrason Sonochem. 2022;82:105903-.
135. Liu J, Ma W, Kou W, Shang L, Huang R, Zhao J. Poly-amino acids coated gold nanorod and doxorubicin for synergistic photodynamic therapy and chemotherapy in ovarian cancer cells. Biosci Rep. 2019;39(12):BSR20192521.
136. Wang L, Wang L, Xu T, Guo C, Liu C, Zhang H, et al. Synthesis of 15P-conjugated PPy-modified gold nanoparticles and their application to photothermal therapy of ovarian cancer. Chemical Research in Chinese Universities. 2014;30(6):959-64.
137. Al-Shammari AM, Al-Saadi H, Al-Shammari SM, Jabir MS. Galangin enhances gold nanoparticles as anti-tumor agents against ovarian cancer cells. 2ND INTERNATIONAL CONFERENCE ON MATERIALS ENGINEERING & SCIENCE (IConMEAS 2019): AIP Publishing; 2020.
138. Piktel E, Oscilowska I, Suprewicz Ł, Depciuch J, Marcińczyk N, Chabielska E, et al. Peanut-Shaped Gold Nanoparticles with Shells of Ceragenin CSA-131 Display the Ability to Inhibit Ovarian Cancer Growth In Vitro and in a Tumor Xenograft Model. Cancers (Basel). 2021;13(21):5424.
139. Ramesh K, Sreenivasan K. Digitonin Conjugated Gold Nanoparticles Show Increased Cytotoxicity to L929 Cells. Journal of Nanopharmaceutics and Drug Delivery. 2013;1(4):388-93.
140. Ding W, Liang Z, El-Kott AF, El-Kenawy AE. Investigation of anti-human ovarian cancer effects of decorated Au nanoparticles on Thymbra spicata extract modified Fe3O4 nanoparticles. Arabian Journal of Chemistry. 2021;14(7):103205.
141. Kotcherlakota R, Srinivasan DJ, Mukherjee S, Haroon MM, Dar GH, Venkatraman U, et al. Engineered fusion protein-loaded gold nanocarriers for targeted co-delivery of doxorubicin and erbB2-siRNA in human epidermal growth factor receptor-2+ ovarian cancer. Journal of Materials Chemistry B. 2017;5(34):7082-98.
142. Kotcherlakota R, Vydiam K, Jeyalakshmi Srinivasan D, Mukherjee S, Roy A, Kuncha M, et al. Restoration of p53 Function in Ovarian Cancer Mediated by Gold Nanoparticle-Based EGFR Targeted Gene Delivery System. ACS Biomaterials Science & Engineering. 2019;5(7):3631-44.
143. Maity R, Chatterjee M, Banerjee A, Das A, Mishra R, Mazumder S, et al. Gold nanoparticle-assisted enhancement in the anti-cancer properties of theaflavin against human ovarian cancer cells. Materials Science and Engineering: C. 2019;104:109909.
144. Bhattacharya R, Patra CR, Earl A, Wang S, Katarya A, Lu L, et al. Attaching folic acid on gold nanoparticles using noncovalent interaction via different polyethylene glycol backbones and targeting of cancer cells. Nanomedicine: Nanotechnology, Biology and Medicine. 2007;3(3):224-38.
145. Van de Broek B, Devoogdt N, D’Hollander A, Gijs H-L, Jans K, Lagae L, et al. Specific Cell Targeting with Nanobody Conjugated Branched Gold Nanoparticles for Photothermal Therapy. ACS Nano. 2011;5(6):4319-28.
146. Chen Y, Xu M, Guo Y, Tu K, Wu W, Wang J, et al. Targeted chimera delivery to ovarian cancer cells by heterogeneous gold magnetic nanoparticle. Nanotechnology. 2016;28(2):025101.
147. Roma-Rodrigues C, Raposo LR, Cabral R, Paradinha F, Baptista PV, Fernandes AR. Tumor Microenvironment Modulation via Gold Nanoparticles Targeting Malicious Exosomes: Implications for Cancer Diagnostics and Therapy. Int J Mol Sci. 2017;18(1):162.
148. Baskar G, Garrick BG, Lalitha K, Chamundeeswari M. Gold nanoparticle mediated delivery of fungal asparaginase against cancer cells. Journal of Drug Delivery Science and Technology. 2018;44:498-504.
149. Bhattacharyya S, Khan JA, Curran GL, Robertson JD, Bhattacharya R, Mukherjee P. Efficient delivery of gold nanoparticles by dual receptor targeting. Adv Mater. 2011;23(43):5034-8.
150. Farooq MU, Novosad V, Rozhkova EA, Wali H, Ali A, Fateh AA, et al. Gold Nanoparticles-enabled Efficient Dual Delivery of Anticancer Therapeutics to HeLa Cells. Sci Rep. 2018;8(1):2907-.
151. Jeon S, Jeon H. Doxorubicin-loaded oligonucleotide conjugated gold nanoparticles: A promising drug delivery system for ovarian cancer. Gynecologic Oncology. 2020;159:117.
152. Tabish TA, Dey P, Mosca S, Salimi M, Palombo F, Matousek P, et al. Smart Gold Nanostructures for Light Mediated Cancer Theranostics: Combining Optical Diagnostics with Photothermal Therapy. Adv Sci (Weinh). 2020;7(15):1903441-.
153. Guo J, Rahme K, He Y, Li L-L, Holmes JD, O’Driscoll CM. Gold nanoparticles enlighten the future of cancer theranostics. Int J Nanomedicine. 2017;12:6131-52.
154.    Li, J., S. Gupta, and C. Li, Research perspectives: gold nanoparticles in cancer theranostics. Quantitative imaging in medicine and surgery, 2013. 3(6): p. 284.
155. Heo DN, Yang DH, Moon H-J, Lee JB, Bae MS, Lee SC, et al. Gold nanoparticles surface-functionalized with paclitaxel drug and biotin receptor as theranostic agents for cancer therapy. Biomaterials. 2012;33(3):856-66.
156. Chen W, Bardhan R, Bartels M, Perez-Torres C, Pautler RG, Halas NJ, et al. A molecularly targeted theranostic probe for ovarian cancer. Mol Cancer Ther. 2010;9(4):1028-38.
157. Same S, Aghanejad A, Akbari Nakhjavani S, Barar J, Omidi Y. Radiolabeled theranostics: magnetic and gold nanoparticles. Bioimpacts. 2016;6(3):169-81.
158. D’Hollander A, Vande Velde G, Jans H, Vanspauwen B, Vermeersch E, Jose J, et al. Assessment of the Theranostic Potential of Gold Nanostars-A Multimodal Imaging and Photothermal Treatment Study. Nanomaterials (Basel). 2020;10(11):2112.
159. Zhu J, Zheng L, Wen S, Tang Y, Shen M, Zhang G, et al. Targeted cancer theranostics using alpha-tocopheryl succinate-conjugated multifunctional dendrimer-entrapped gold nanoparticles. Biomaterials. 2014;35(26):7635-46.
160. Jokerst JV, Cole AJ, Van de Sompel D, Gambhir SS. Gold nanorods for ovarian cancer detection with photoacoustic imaging and resection guidance via Raman imaging in living mice. ACS nano. 2012;6(11):10366-77.
161. Bassi B, Dacarro G, Galinetto P, Giulotto E, Marchesi N, Pallavicini P, et al. Tailored coating of gold nanostars: rational approach to prototype of theranostic device based on SERS and photothermal effects at ultralow irradiance. Nanotechnology. 2018;29(23):235301.
162. Hajebi S, Rabiee N, Bagherzadeh M, Ahmadi S, Rabiee M, Roghani-Mamaqani H, et al. Stimulus-responsive polymeric nanogels as smart drug delivery systems. Acta Biomater. 2019;92:1-18.
163. Jindal M, Nagpal M, Singh M, Aggarwal G, Dhingra GA. Gold Nanoparticles- Boon in Cancer Theranostics. Current Pharmaceutical Design. 2020;26(40):5134-51.
164. Zafar A, Alruwaili NK, Imam SS, Alharbi KS, Afzal M, Alotaibi NH, et al. Novel nanotechnology approaches for diagnosis and therapy of breast, ovarian and cervical cancer in female: A review. Journal of Drug Delivery Science and Technology. 2021;61:102198.
165. Liang H, Tian H, Deng M, Chen X. Gold Nanoparticles for Cancer Theranostics. Chinese Journal of Chemistry. 2015;33(9):1001-10.
166. Almeida JPM, Figueroa ER, Drezek RA. Gold nanoparticle mediated cancer immunotherapy. Nanomedicine. 2014;10(3):503-14.
167. Dreaden EC, Alkilany AM, Huang X, Murphy CJ, El-Sayed MA. The golden age: gold nanoparticles for biomedicine. Chem Soc Rev. 2012;41(7):2740-79.
168. Bailly A-L, Correard F, Popov A, Tselikov G, Chaspoul F, Appay R, et al. In vivo evaluation of safety, biodistribution and pharmacokinetics of laser-synthesized gold nanoparticles. Sci Rep. 2019;9(1):12890-.