Translate this page into:
Dendritic cell engineering for selective targeting of female reproductive tract cancers
For correspondence: Dr Pradyumna Kumar Mishra, Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Kamla Nehru Hospital Building (Gandhi Medical College Campus), Bhopal 462 001, Madhya Pradesh, India e-mail: pkm_8bh@yahoo.co.uk
-
Received: ,
This is an open access journal, and articles are distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as appropriate credit is given and the new creations are licensed under the identical terms.
This article was originally published by Wolters Kluwer - Medknow and was migrated to Scientific Scholar after the change of Publisher.
Abstract
Female reproductive tract cancers (FRCs) are considered as one of the most frequently occurring malignancies and a foremost cause of death among women. The late-stage diagnosis and limited clinical effectiveness of currently available mainstay therapies, primarily due to the developed drug resistance properties of tumour cells, further increase disease severity. In the past decade, dendritic cell (DC)-based immunotherapy has shown remarkable success and appeared as a feasible therapeutic alternative to treat several malignancies, including FRCs. Importantly, the clinical efficacy of this therapy is shown to be restricted by the established immunosuppressive tumour microenvironment. However, combining nanoengineered approaches can significantly assist DCs to overcome this tumour-induced immune tolerance. The prolonged release of nanoencapsulated tumour antigens helps improve the ability of DC-based therapeutics to selectively target and remove residual tumour cells. Incorporation of surface ligands and co-adjuvants may further aid DC targeting (in vivo) to overcome the issues associated with the short DC lifespan, immunosuppression and imprecise uptake. We herein briefly discuss the necessity and progress of DC-based therapeutics in FRCs. The review also sheds lights on the future challenges to design and develop clinically effective nanoparticles-DC combinations that can induce efficient anti-tumour immune responses and prolong patients’ survival.
Keywords
Cancer therapy
dendritic cells
nanomedicine
translational oncology
Introduction
Cancer accounted for approximately 9.6 million deaths and 18.1 million new cases globally in 20181. Among all female reproductive tract cancers (FRCs), the most common are the cancers of the cervix (cervical cancer), ovary and endometrium, which contribute to more than three-fourth of the total reported cases. Cervical cancer is the third most common malignancy in women which resulted in almost 300,000 deaths worldwide in 2018 (about 7.5% of all deaths from cancer in females)1. The overall mortality to incidence ratio of cervical cancer is almost 50 per cent due to which it is a major problem in developing and underdeveloped countries12. Other frequently occurring FRCs also display a similar scenario, as cancers of endometrium and ovary, respectively, stand sixth and seventh most frequently occurring malignancies in women worldwide. Approximately, 382,069 and 295,414 new cases of endometrial and ovarian cancers, respectively, were reported worldwide in 2018. The occurrence rate of endometrial cancer in developing countries is more than three times to cervical cancer1234. Despite several efforts, stage-dependent therapeutic response is considered as a major reason for the higher mortality rate observed in these cancers. This creates a pressing need to develop novel approaches for the prevention and treatment of FRCs.
Standard therapeutics and their limitations
The late detection and limited therapeutic options available for FRCs significantly contribute to the global rise in mortality due to these malignancies. Similar to other malignancies, the primary therapy of FRCs is the surgical tumour removal which may be used alone or with other therapies (radio- or chemotherapy). Occasionally, hormone therapy such as progestin (a synthetic drug similar to the hormone progesterone) is also administered. In general, the treatment option mainly relies on the tumour type, occurrence site and disease phase56. For instance, in endometrial cancers, radiation therapy may be given in combination to surgery if the disease is malignant and spreading to the nearby cervix, ovary or lymph nodes, which results in a better prognosis. Chemotherapeutic drugs such as carboplatin, cisplatin, doxorubicin and paclitaxel remain the choice of treatment if the disease is recurring or spreading beyond the uterus and cervix. These standard therapies have been shown to reduce the tumour size and control its spread; however, recurrence-induced toxicity and side effects remain severe issues78. Furthermore, a large number of female reproductive tract tumours metastasize before disease identification which significantly reduces the effectiveness of these traditional procedures9. For example, in cervical cancer, the disease starts as the growth of pre-cancerous lesions in the cervix region (stage I) and extends within pelvic cavity, side wall or lower vagina (stages II and III) and other body parts (stage IV)10. The prognosis after treatment gets even worse with increasing stages for advanced cancer; the cure rate (5 year survival) drops from about 45 per cent for earlier stages to as low as 15 per cent in advanced stages1112. Due to various reasons including unawareness and fear, >80 per cent of affected population seeks help at later stages, thus worsening the disease outcomes. Moreover, the restricted response to high-dose standard therapies observed among different gynaecological malignancies increases the probability of disease recurrence as resistant disease. The unavailability of systemic therapies causes major harm to other normally growing cells of different vital systems including immune system, which may lead to an immunocompromised state, thereby increasing the probability of patients to develop various opportunistic infections13.
Immunotherapeutics: An alternate strategy
Successful treatment of FRCs remains a major challenge and requires urgent attention for developing novel therapeutic strategies. In the past decade, significant efforts have been made to utilize immune cells and mechanisms to remove unwanted tumour cells. Normally, different components of immune system comprehensively check and remove unnecessary cells and other entities through a tightly regulated mechanism known as immune surveillance. This involves innate and adaptive immune responses which work under strict coordination to generate anti-tumour responses. The key for the generation of an immunotherapeutic strategy lies within these responses as these can be induced or modulated through a number of immunological agents. These strategies mainly depend on three basic strategies, i.e. increasing processing and presentation of tumour antigens (Ag), modulating T-cell responses and overcoming cancer cell-induced immunosuppression (Fig. 1). These strategies have been widely utilized to develop an effective anti-cancer therapy for gynaecological malignancies1415. Among the various analyzed immune-based strategies, utilizing the ability of dendritic cells (DCs) to initiate immune responses makes these cells as appropriate therapeutic alternate for FRCs. Antigen presenting cells (APCs) such as DCs are one of the important constituents of immune mechanisms and possess the potential of being utilized for anti-cancer therapeutics. The superior capability of DCs to uptake, process and present foreign antigens holds optimum promise for utilizing these cells for developing a successful supportive care for gynaecological malignancies.

- An overview of widely used immune targets for cancer therapeutics. Different strategies utilizing antigen presenting cells, T-lymphocytes, macrophages and specific antibodies to specifically target tumour cells. CD, cluster of differentiation; DCs, dendritic cells; IL, interleukin; MHC, major histocompatibility complex; PD1, programmed cell death protein 1; TCR, T-cell receptor; IDO, indoleamine-pyrrole 2,3-dioxygenase.
Dendritic cell immunotherapeutics for female reproductive tract cancers
DC, initially identified in 1973 by the pioneer work of R.M. Steinman and Z.A. Cohn, are the professional APCs of our body. These cells possess a significant potential to initiate primary immunological responses. DCs identify and interact with foreign molecules through their pattern-recognition receptors to release different immunological mediators and trigger effective host immune responses16. This centralized role of antigen processing and presentation through specialized surface receptors makes DCs a key player in initiating and regulating responses against tumour cells. This vital role of DCs to regulate anti-tumour responses is being broadly utilized towards developing personalized cancer immunotherapeutics (Fig. 2). DCs take up foreign antigen, process through specialized major histocompatibility (MHC) complexes and present these processed antigen to T-cells for the generation of efficient immunological response. However, this also requires the appropriate presentation of different co-stimulatory and signalling molecules. Normally, after uptake, antigens are processed either via endogenous pathway in which intracellular antigens are processed through class-I MHC or by exogenous pathway whereas extracellular antigens are presented with MHC-II17. In endogenous pathway, the antigens are directed to proteasomal degradation through ubiquitination which helps them to fit in the peptide-binding region of MHC class-I. These peptides bind with a protein known as transport-associated protein-1 and 2 heterodimer, which aids their transfer to rough endoplasmic reticulum (ER)18. These peptides are co-presented with MHC-I on the DCs membrane. However, in exogenous pathway, the endocytosed antigens are degraded by endosomal proteases which then fuses with MHC class-II in the rough ER with the help of HLA-DM and the stable peptide-MHC composite is then presented on DCs18. The DCs also possess capability to cross-present exogenous antigens with class-I MHC to persuade a Th1-mediated response. In this pathway, the cells initiate antigen uptake in exogenous mode but later switches to the endogenous pathway. It involves the retrotranslocation of endosomal compartments and proteasomal complex to load exogenous antigens on class-I MHC19. This ability of DCs is important and has been widely assessed to develop a competent DC-based immunotherapy against FRCs.

- A diagrammatic representation of developing dendritic cell vaccines for female reproductive tract cancers. Dendritic cells are isolated from peripheral blood and cultured with growth cytokines. These cultured dendritic cells are then pulsed with appropriate tumour antigens and administered to the patients. IL, interleukin; GM-CSF, granulocyte macrophage-colony stimulating factor; Ag, antigen.
Clinical efficiency and impediments
DC-based therapeutic vaccines for FRCs has undergone a series of in vitro examinations and now reached the stage of preclinical and clinical trials. A set of clinical considerations, including host- and tumour-related factors such as age, immunosuppression, stage of disease, HLA and co-infection have been recognized by the Cancer Vaccine Clinical Trial Working Group to develop a successful DC vaccine20. It is also vital to have a standardized protocol for optimum DC culture and vaccination. Several ongoing and completed clinical studies have successfully established the clinical efficiency of DC-based vaccines in FRCs (Table I). A series of investigations conducted with the peptide-pulsed DCs has shown the generation of significant anti-tumour responses without any significant toxicity3233. In one such phase II investigation among patients of ovarian cancer with elevated recurrence risk, it was reported that p53 peptide-pulsed DC vaccines were safe and efficient in generating specific responses against p53 peptide23. In another phase I trial, therapeutic administration of DCs pulsed with the recombinant HER-2-granulocyte macrophage-colony stimulating factor (GMCSF) peptide blend to a group of patients comprising metastatic ovarian cancer was observed to generate a strong anti-HER-2 response21. Studies have also shown keyhole limpet haemocyanin (KLH) act as a vital antigen among different gynaecological malignancies. A phase I study conducted on patients suffering from uterine and ovarian cancers suggested that administration of KLH and whole T-lymphocytes (TLs) (autologous) activated DCs was safe and immunologically active22. Similarly, subcutaneous (s.c.) vaccination of KLH-loaded DCs to the patients resulted in significant induction of protein-specific cellular immune responses and delayed-type hypersensitivity reactions, which reaffirmed the safety and effectiveness of these vaccines24.
Cancer | Strategy used | Reference |
---|---|---|
Ovarian | DCs loaded with peptides of MUC1/HER-2/NEU | 21 |
Ovarian and uterine | DC pulsed with KLH and autologous tumour antigens | 22 |
Ovarian | DCs pulsed with p53 peptide along with IL2 | 23 |
Ovarian | Chemotherapy followed by doses of TL-KLH co-loaded pulsed DCs followed by IL2 dosage as an immune adjuvant | 24 |
Cervical | HPV protein loaded DC | 25 |
Ovarian | DCs loaded with HER-2/NEU, hTERT, and PADRE peptides, with or without lowdose intravenous cyclophosphamide | 26 |
Ovarian | TL co-incubated DCs | 27 |
Uterine | DCs loaded with WT1-mRNA | 28 |
Ovarian | DCs loaded with oxidized TL along with bevacizumab and cyclophosphamide | 29 |
Ovarian and cervical | Autologous DC formulation | 30 |
Cervical | TL primed DC followed by cisplatin | 31 |
MUC1, mucin 1; HER-2, herceptin-2; KLH, keyhole limpet haemocyanin; DC, dendritic cells; TL, tumour lysate; HPV, human papiloma virus; IL, interleukin; h-TERT, telomerase reverse transcriptase; PADRE, pan HLA DR-binding epitope; WT1, wilms tumour 1
To validate the clinical efficacy of this adjuvant therapy, the vaccines have been systematically administered and assessed in different combinatorial modes. In a randomized phase I trial, loaded DCs along with a chemotherapy drug cisplatin were administered to cervical cancer patients. Absolute clinical response with no recurrence for five years was reported in one of three individuals with the metastatic disease which suggested that the strategy might act as appropriate adjuvant therapy for advanced cervical cancers31. Similarly, a trial conducted on patients with recurrent ovarian cancers further reaffirmed the clinical efficiency of these vaccines. The study utilized DCs co-incubated with oxidized TL together with a chemotherapy drug, bevacizumab and observed strong anti-tumour response among the treated patients which later strongly correlated with the disease stability29. In one randomized trial vaccination of HER-2 + telomerase reverse transcriptase (h-TERT) + pan HLA DR-binding epitope (PADRE) peptides, pulsed DCs in combination with cyclophosphamide to the advanced ovarian cancer patients resulted in elicited tumour cell-specific responses with promising survival ability26. In a different strategy, treatment of WT1 mRNA electroporated DCs to ovarian cancer patients showed higher anti-tumour immunological responses which also correlated with the extended patient survival28. In the past decade, these clinical trials have shown promising results and suggested that the establishment of a successful DC-based approach requires utilization of synergistic therapy combinations and incorporation of novel modalities which can generate profound anti-tumour responses and prolong patients’ survival in FRCs.
Immunosuppressive microenvironment
The efficiency of DC-based vaccines against FRCs has been assessed in several pre-clinical and clinical studies; however, the key problem in the clinical triumph of DC immunotherapy is the well-established immunosuppressive surroundings. These tolerogenic surroundings limit the immune activation ability of DCs and result in a limited therapeutic response34. Different mechanisms have been described which help tumour cells to manipulate host immune system and escape surveillance for progression. These mechanisms primarily aim to downregulate the expression of tumour antigens and MHC molecules, facilitating cancer cells to get away from infiltrating T-cells34. The immunosuppressive tumour cell surroundings primarily comprise numerous suppressive molecules and cells which jointly work to create a tolerogenic microenvironment35. These molecules suppress the secretion of inflammatory molecules and assist in recruiting Treg populations to downregulate the activity of T-cell subsets, natural killer cells and DCs35. Although exact molecular pathways and their inter-linking mechanisms are still a matter of investigation, evidence signifies the vital function of tumour growth factor (TGF)-β in tumour development and its metastatic progression36. Interleukin (IL)-10 has also been observed to significantly limit the appearance of different co-stimulatory molecules and inhibit the vital process of antigen processing and presentation37. T-cells interacting with IL-10 expressing DCs are shown to be functionally anergic38. Molecules such as vascular endothelial growth factor-A (VEGF-A) and prostaglandin E2 (PGE2) interfere with nuclear factor-κB, G protein-coupled receptor and other transcription factors to negatively influence DC maturation and function3940. Moreover, PGE2 is also shown to influence the levels of an immunoregulatory enzyme, indoleamine-2,3-dioxygenase (IDO) which later suppresses the anti-tumour responses4142. Tumours also release soluble Wnt ligands to actively tolerize DC populations within the tumour microenvironment. It has also been shown that the involvement of Wnt-β-catenin pathways have a vital role in suppression of pro-inflammatory cytokines and elevated expression of the immunosuppressive cytokines43. Different studies have shown that this tumour-associated immunosuppression is significantly linked to the aggressiveness and metastasis of different cancers of the female reproductive tract. Patients with higher TGF expression are more closely related to the worse prognosis of post-surgical therapeutic interventions44. Similarly, differential expression of cell surface molecules such as intercellular adhesion molecule and MHC on tumour cells supports their escape from DC vaccine-generated specific immune responses4546. Therefore, to develop clinically effective DC-based cancer therapeutics, it is important to design strategies which can potentially counter this immunosuppressive microenvironment.
Tumour-initiating cells
Tumour-initiating cells (TRICs) have a key function in developing therapeutic tolerance and progression of FRCs. This dedicated cell population functions as tumour stem cells and shows higher tendency for metastasis and increased therapeutic resistance. However, the exact mechanisms depicting the development of such resistive abilities of TRICs are largely unknown. It is assumed that the reciprocal crosstalk between the malignant cells and tumour-infiltrating leukocytes probably regulates the expansion of stem cell-like population and facilitates developing therapeutic resistance among these cells47. TRICs express membrane efflux transporters which further support their chemoresistance characteristics46. In addition, enhanced DNA repair and low mitotic index are other properties which contribute to the development of drug resistance. Further, these self-renewing malignant progenitors form a group of cells to regulate cellular differentiation47. TRICs are the major cause of clinical relapse as they escape the therapeutics and re-grow upon their cessation and can subsequently regenerate entire malignant phenotype484950. The well-known immunosuppressive tumour microenvironment in patients and significant toxicity of traditional therapeutics further assist TRICs to escape the immunological surveillance50. This represents a decisive interface between vaccine effectiveness and tumour progression, so it is vital to devise and include new clinically effective approaches enabling DCs to overcome tumour-induced immunological suppression along with eliciting specific immune responses for removal of TRICs, a major cause of clinical relapse in gynaecological malignancies.
Dendritic cell nanoengineering: An approach to move forward
Nanotherapeutics offers an efficient inclusion in traditional DC-based vaccine to surpass the existing obstacle of immune suppressive microenvironment51. It has been shown that tumour antigens have limited ability to activate DCs; thus, co-encapsulating tumour antigens with different adjuvants such as micro-emulsions, lipopolysaccharides and bacterial CpG DNA may prove beneficial51 (Fig. 3). Encapsulation of tumour antigens using different nanomaterials protects them from damage of proteases and assists in a prolonged and proscribed antigenic release to DCs. This increases the antigenic acquaintance to DCs and helps them to counter the suppressive microenvironment34. Incorporation of different surface modifiers and ligands may specifically direct nanocarriers to target cells and further aid the clinical and immune activation potential of DC vaccines. Earlier reports have shown that encapsulation of tumour antigens enhances the MHC-I-mediated antigen cross-presentation3450. However, for efficient immune stimulation, different factors such as size, shape, surface chemistry, mode of administration, diffusion and solubility play an important role5253. Evidence suggests that reduction in the ionic interaction of matrix material with the encapsulated protein significantly declines the burst effect and reduces the release rate54. Therefore, copolymers such as ethylene oxide-propylene oxide can be incorporated to a non-toxic polymer with appropriate molecular weight and chemically defined monomers to improve the efficiency of nanocarriers54.

- Outline of using nanoencapsulated tumour antigens for dendritic cell engineering. The tumour antigens can be initially encapsulated and then utilized for pulsing the dendritic cells. These tumour antigen presenting dendritic cells can be administered to the patients. NPs, nanoparticles.
Vital factors for dendritic cell nanoengineering
The therapeutic efficiency of DC vaccines can be further modulated by the mode of uptake by DCs, which have a primary function in shaping the antigenic fate, i.e. either interact with MHC-I or with MHC-II. However, relying on the physical and chemical compositions of the encapsulated Ag the mode of uptake may be endocytosis, macropinocytosis and phagocytosis. A pre-requisite for successful DC-based therapeutic vaccines is their ability to generate a potent Th1-mediated cytotoxic response. It is suggested that larger nanoparticles (NPs) with size approximately 500 nm are preferably taken up through phagocytosis, a process which assists humoral immune responses55. The NPs with a smaller size (approximately 100 nm) are effectively taken up through endocytosis which generates a potent Th1 response56. Thus, the use of NPs with size less than 100 nm should be preferred to ensure the generation of an optimum cytotoxic immune response. The larger particles (500 nm) predominantly reside at the injection site while the smaller ones freely move to the lymph node area where these interact with resident DCs. Smaller particles also provide higher surface area and superior membrane permeability which support their efficient uptake57. A quick particle uptake and longer post-injection retention of smaller particles (<100 nm) have also been reported58. This suggests the suitability of these particles for designing future in vivo targeting strategies. In addition, maintaining the appropriate particle shape is also crucial as spherical particles are supposed to have less premature clearance, more circulatory halftime and good organ distribution5859.
Following size and shape of the particle, another vital factor for developing effective nanoengineered DC vaccines is the mode of administration. NPs injected through intraperitoneal mode interact with the macrophages, while, upon intradermal (i.d.) administration, NPs are taken up by DCs. It has been reported that i.d. injected particles move preferentially to lymphatics with an average 50 per cent higher bioavailability as compared to intramuscular (i.m.) mode59. Moreover, the kinetics and magnitude of tumour cell-specific Th1 responses were reported to be strongly influenced by the mode of administration [i.d., i.m., intralymphatic (i.l.) and s.c.]. Administration of different NPs such as chitosan, liposomes and poly(lactic-co-glycolic acid) (PLGA) through i.l. route induces a strong Th1-type immunological response which is a determining factor for therapeutic use of nanoengineered DCs60.
Nanocarriers for dendritic cell nanoengineering
Different nanocarrier systems have been evaluated for the treatment of FRCs. These nanocarriers including lipid-based and/or polymer-based systems have been broadly used to deliver therapeutic drugs/tumour antigens and have also shown to possess the ability to target both tumour cells and microenvironment. The HPV16 E7 small interfering (si) RNA-loaded chitosan NPs were reported to successfully restrict the tumour cell proliferation and reverse the drug resistance of ovarian tumour cells, suggesting these nanocarriers as promising therapeutic mediators for treating gynaecological malignancies61. Similarly, cisplatin-loaded liposomal formulations effectively targeted cancer cells of the female reproductive tract. The observed growth inhibitory effects were better than that of free cisplatin62. The liposomal formulation comprising a new T7 peptide which exclusively attaches to the transferrin receptor, reported best in vivo growth inhibitory effect on ovarian carcinoma cells63. In addition, in comparison to the plain liposomal doxorubicin, peptide-conjugated liposomal doxorubicin was shown to be better in controlling tumours64.
Polymeric NPs such as PLGA are also effectively analyzed for the release of therapeutic molecules. The delivery of tumour-associated antigens through PLGA-NPs enables DCs to proficiently trigger tumour-specific responses. The encapsulation angiogenesis inhibitors (TNP-470-NP-APRPG) through PLGA showed promising results for utilizing the strategy for treating ovarian cancers65. Administration of short hairpin (sh) RNA encapsulated with PLGA showed significantly higher anti-tumour effects against ovarian cancer cells66. Therapeutic efficiency of novel folic acid decorated-PEG-PLGA NPs for targeting the drugs to cancer cells in endometrial cancers has also been shown67. The dosage of PLGA-PRINT NPs loaded with chemotherapeutic drug docetaxel and chitosan NPs containing mEZH2 siRNA was reported to be associated with considerable anti-angiogenic and pro-apoptotic consequences in tumour cells68. NPs carrying oligonucleotide duplexes dramatically raised the immune stimulatory activity of miR-155 in DCs related to ovarian cancers69. It has been shown that surface-modified PEG-poly caprolactin-NPs strongly activate DCs for antigen cross-presentation and thus can be used as a potent immune adjuvant70.
A number of other NPs have been used for directing immune cells to target and increase the eradication of gynaecological tract tumours. Recently, the combination of NPs with albumin-bound paclitaxel and nedaplatin was observed to be active and well tolerable for treating cervical cancer patients with late-stage, frequent or metastatic disease70. Carbon NPs were shown to precisely envisage the pathological status of pelvic lymph nodes in early cervical tumours, suggesting their larger function towards generating DC immunotherapy against these cancers71. Synergistic combination of micellar-based telodendrimer nanocarrier systems with chemotherapeutic drugs paclitaxel and cisplatin resulted in a competent and targeted delivery of drug to tumour cells along with reduced toxicity and potent immunological effect72. The efficiency of this system has been re-validated with bortezomib and doxorubicin where these multifunctional telodendrimer nanocarriers were reported to restore therapeutic synergy with minimum cytotoxicity in ovarian cancers73. PEGylated peptide diaminocyclohexyl-platinum conjugates are suggested to be new prospective drug release method with enhanced anti-tumour efficacy and clinical possibility in treatment of ovarian cancer malignancies74. The superiority of cationic liposomes and Toll-like receptor (TLR)-3 agonist complexes towards improving the tumour-specific immune response has been attributed to the TLR3-interferon regulatory factor signalling within DCs75.
Several NP formulations including polymeric formulations and lipid-based systems have been utilized for developing a successful nanocarrier system for efficient antigen delivery to DCs. Among the studied NPs, solid-lipid NPs (SLN) offer the most efficient delivery system. SLN systems initially prepared in the early 1990s belong to class of the standard colloidal systems which have a solid-lipid core making internal core minimally exposed to water, thereby possessing increased stability of the loaded material76. Besides, the surface of these NPs can be simply attached with appropriate ligands for targeting, has superior payload and is minimal toxic52. SLN is prepared by utilizing lipids such as triglycerides which have high melting points and their lipid core is stabilized by different emulsifiers. In addition, the ease of surface modification enables and ability to co-encapsulate adjuvants further improve their capability to activate DCs and provoke a strong anti-tumour response. These properties have been significantly supported by the reports which suggest SLN as an optimum nano-carrier system7778. Cationic SLN loaded with STAT3 decoy oligodeoxynucleotides was reported to efficiently transfect genes, inhibit invasion and stimulate apoptosis in ovarian cancer cells79. Wang et al79 suggested that SLN surface modified with hyaluronic acid act as a potential carrier to target tumour cells and overcome multidrug resistance associated with in cervical cancers. These studies provide a platform to utilize nanocarrier systems for developing optimum DC-based vaccines. However, the choice of an optimum system is vital but confusing. In a study, a relative evaluation of lipophilic NPs was performed to recognize and develop an optimum NP-based strategy for DC-based targeting of TRICS in gynaecological cancers. The study reported that in comparison to other studied lipid-based nanocarriers, mannosylated-SLN was a feasible and optimum approach. Mannosylated SLN-pulsed DCs were observed to be efficiently taken up by DCs, induced minimal toxicity and were more potent inducers of tumour cell-specific immune responses50.
Nanocarriers for in vivo targeting
The limited existence of the administered DCs restricts their activity and not many of these reach to the lymph nodes which is a major reason for the observed limited effectiveness of these vaccines45. Therefore, focusing on the DCs within the body instead of culturing them ex vivo may be an option. This can be done using NP-mediated DC-targeting approaches which consist of a blend of antigens and other vaccine constituents. Further, addition of immune activating adjuvants may help generate a surrounding environment which aid effective DC recruitment and activation to increase the vaccine efficiency80. In addition, surface modification of the NPs with ligands specific for DC membrane receptors enable the NPs to dynamically target DC with no or minimal non-specific uptake818283. Upon administration, the NPs interact with the in vivo residing DCs, and due to the prolonged antigenic release, these cells come out of suppressive microenvironment and trigger tumour cell-specific immune responses848586.
Earlier evidence also suggests that in vivo DCs targeting is an efficient option with huge immunological prospective878889. In this regard, C-type lectin receptors (CLR) is one of the vital ligands analyzed for precise DC targeting. CLR holds the ability to bind with the antigenic carbohydrates residues. Broadly, CLR is divided into two categories, i.e. type I and type II. Targeting DCs with DEC-205 and CD11c labelled liposomes showed significantly increased tumour-cell specific immune responses90. Perhaps preparing the blend of MHC-I and T-helper cell epitopes with CLR-specific molecules on NPs might hold as a promising approach for inducing a competent tumour cell-specific T-cell responses56. Similarly, liposomes surface modified with glycans exhibited an enhanced encounter with DCs91. Incorporating LPS additionally augmented T-cell activation signifying the anti-tumour potential of the approach91. Upon administration of the NPs (gold nanocarriers and liposomes) modified with Fc receptor, efficient interaction with DC receptor was observed which suggested their potential ability to target DCs and activating tumour cell-specific immune responses92. Investigations have also shown the ability of superoxide iron NPs towards DC targeting and inducing efficient migration in vivo, with superior bio-compatibility9394. This ability of NPs can be further improved by including imaging mediators which enable simultaneous monitoring of nanocarriers and assist in further designing the improved targeted approaches81. Developing such DC targeting strategies will help towards overcoming immune suppressive tumour surroundings and designing efficient strategies for cancer therapy in future. A brief detail of different NPs which can be used for generating nanoengineered DCs is provided in Table II.
Nanoparticle | Properties | References |
---|---|---|
Liposomes | Non-toxic, biocompatible, biodegradable NP | 5195 |
Prompt RES clearance | ||
Approved by FDA for clinical use | ||
Solid lipid nanoparticles | Non-toxic, biocompatible and biodegradable NP | 325096 |
Long-term stable storage | ||
Prompt surface modification | ||
FDA approved for clinical use | ||
Ease in higher pharmaceutical manufacturing | ||
Poly‑(lactic‑co‑glycolic acid) | Non-toxic, biocompatible, biodegradable NP | 9798 |
Long-term stable storage | ||
FDA approved for clinical use | ||
Ease in higher pharmaceutical manufacturing | ||
Poly‑ε‑caprolactone | Non-toxic, biocompatible, biodegradable NP | 99,100 |
Poly (propylene sulphide) | Non-toxic, biocompatible and biodegradable NP | 101 |
Long-term stable storage | ||
Poly (γ‑glutamic acid) nanoparticles | Biocompatible and biodegradable NP | 102 |
Long-term stable storage | ||
Chitosan | Non-toxic, biocompatible, biodegradable NP | 103104 |
Long-term stable storage | ||
Ease in surface modification | ||
Gelatin | Non-toxic, biocompatible, biodegradable NP | 105 |
Trouble-free manufacture | ||
Prompt surface modification |
NP, nanoparticles; RES, reticular endothelial system; FDA, Food and Drug Administration
Conclusion
DCs vaccines offer a potential therapeutic modality for FRCs. The promising results observed in pre-clinical and clinical studies encourage continuing efforts towards further optimizing these methodologies and validating combinatorial therapeutic approaches. A major effort must be dedicated towards designing approaches to overwhelm the well-identified issue of tumour-associated immunological suppression. In this regard, incorporation of nanoengineering approaches may prove vital as these possess the ability to enhance the immune activation property of DCs. These engineered DCs are suggested to be safe and hold significant potential to eradicate TRICs. Encapsulation helps DCs to overcome immune suppression which later initiates a cascade of tumour-specific immune responses. Designing multifunctional NPs may further open the opportunities of direct in vivo targeting and may be vital as it will help in overcoming some important concerns related to ex vivo tumour antigen loading, short life-span of loaded DCs and faulty migration. In addition, the presence of substantial amount of resident DCs in the lymph nodes will assist in the efficient antigen uptake, processing and its cross-presentation to the neighbouring T-cell population to generate specific immune responses. These nanotherapeutic approaches have profoundly worked under different in vitro and in vivo conditions, however; their successful clinical translation relies on the ability of these vaccines to overcome different limiting factors. The present article was an attempt to showcase the need and significance of developing nanoengineered approaches, likely to maximize the therapeutic success of DCs against different FRCs.
Financial support & sponsorship:
Authors thank the Indian Council of Medical Research and Department of Science and Technology, Government of India, New Delhi, for providing necessary financial support.
Conflicts of Interest:
None.
References
- Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394-424.
- [Google Scholar]
- Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136:E359-86.
- [Google Scholar]
- Endometrial adenocarcinoma: Clinicopathologic and survival characteristics in Yazd, Iran. Asian Pac J Cancer Prev. 2014;15:2797-801.
- [Google Scholar]
- National Cancer Intelligence Network. 2008. Cancer incidence and mortality by cancer network, UK, 2005. London: National Cancer Intelligence Network; Available from: http:// www.ncin.org.uk
- [Google Scholar]
- Changing pathology with changing drugs: Tumors of the gastrointestinal tract. Pathobiology. 2011;78:76-89.
- [Google Scholar]
- Factors prognostic of survival in advanced-stage uterine serous carcinoma. Gynecol Oncol. 2017;146:27-33.
- [Google Scholar]
- Race and hormone receptor-positive breast cancer outcomes in a randomized chemotherapy trial. J Natl Cancer Inst. 2012;104:406-14.
- [Google Scholar]
- Reciprocal links between venous thromboembolism, coagulation factors and ovarian cancer progression. Thromb Res. 2017;150:8-18.
- [Google Scholar]
- Recurring gastrointestinal stromal tumor with splenic metastasis. J Korean Surg Soc. 2011;81(Suppl 1):S25-9.
- [Google Scholar]
- Cervical cancer: Biomarkers for diagnosis and treatment. Clin Chim Acta. 2015;445:7-11.
- [Google Scholar]
- Epidemiology of epithelial ovarian cancer. Best Pract Res Clin Obstet Gynaecol. 2017;41:3-14.
- [Google Scholar]
- Optimizing treatment in recurrent epithelial ovarian cancer. Expert Rev Anticancer Ther. 2017;17:1147-58.
- [Google Scholar]
- Immunosuppression in patients with high-grade gliomas treated with radiation and temozolomide. Clin Cancer Res. 2011;17:5473-80.
- [Google Scholar]
- A cancer vaccine with dendritic cells differentiated with GM-CSF and IFNα and pulsed with a squaric acid treated cell lysate improves T cell priming and tumor growth control in a mouse model. Bioimpacts. 2018;8:211-21.
- [Google Scholar]
- Cytokine-induced memory-like natural killer cells have enhanced function, proliferation, and in vivo expansion against ovarian cancer cells. Gynecol Oncol. 2019;153:149-57.
- [Google Scholar]
- The response of human dendritic cells to co-ligation of pattern-recognition receptors. Immunol Res. 2012;52:20-33.
- [Google Scholar]
- Trial watch: Dendritic cell-based anticancer immunotherapy. Oncoimmunology. 2017;6:e1328341.
- [Google Scholar]
- A detailed analysis of the murine TAP transporter substrate specificity. PLoS One. 2008;3:e2402.
- [Google Scholar]
- The role of cDC1s in vivo: CD8 T cell priming through cross-presentation. F1000Res. 2017;6:98.
- [Google Scholar]
- Clinical considerations in developing dendritic cell vaccine based immunotherapy protocols in cancer. Curr Mol Med. 2009;9:725-31.
- [Google Scholar]
- Induction of cytotoxic T-lymphocyte responses in vivo after vaccinations with peptide-pulsed dendritic cells. Blood. 2000;96:3102-8.
- [Google Scholar]
- Vaccination with autologous tumour antigen-pulsed dendritic cells in advanced gynaecological malignancies: Clinical and immunological evaluation of a phase I trial. Cancer Immunol Immunother. 2002;51:45-52.
- [Google Scholar]
- A gynecologic oncology group phase II trial of two p53 peptide vaccine approaches: Subcutaneous injection and intravenous pulsed dendritic cells in high recurrence risk ovarian cancer patients. Cancer Immunol Immunother. 2012;61:373-84.
- [Google Scholar]
- Therapeutic DC vaccination with IL-2 as a consolidation therapy for ovarian cancer patients: a phase I/II trial. Cell Mol Immunol. 2015;12:87-95.
- [Google Scholar]
- Human papillomavirus type 16 and 18 E7-pulsed dendritic cell vaccination of stage IB or IIA cervical cancer patients: A phase I escalating-dose trial. J Virol. 2008;82:1968-79.
- [Google Scholar]
- Phase I/II randomized trial of dendritic cell vaccination with or without cyclophosphamide for consolidation therapy of advanced ovarian cancer in first or second remission. Cancer Immunol Immunother. 2012;61:629-41.
- [Google Scholar]
- Phase I/II clinical trial of a Wilms’ tumor 1-targeted dendritic cell vaccination-based immunotherapy in patients with advanced cancer. Cancer Immunol Immunother. 2019;68:121-30.
- [Google Scholar]
- Immunological response after WT1 mRNA-loaded dendritic cell immunotherapy in ovarian carcinoma and carcinosarcoma. Anticancer Res. 2013;33:3855-9.
- [Google Scholar]
- A phase I vaccine trial using dendritic cells pulsed with autologous oxidized lysate for recurrent ovarian cancer. J Transl Med. 2013;11:149.
- [Google Scholar]
- Development and clinical evaluation of dendritic cell vaccines for HPV related cervical cancer - a feasibility study. Asian Pac J Cancer Prev. 2014;15:5909-16.
- [Google Scholar]
- Effectiveness of immune therapy combined with chemotherapy on the immune function and recurrence rate of cervical cancer. Exp Ther Med. 2015;9:1063-7.
- [Google Scholar]
- Assessment of tumor antigen-loaded solid lipid nanoparticles as an efficient delivery system for dendritic cell engineering. Nanomedicine (Lond). 2013;8:1067-84.
- [Google Scholar]
- A phase I trial of immunotherapy with lapuleucel-T (APC8024) in patients with refractory metastatic tumors that express HER-2/neu. Clin Cancer Res. 2009;15:5937-44.
- [Google Scholar]
- Dendritic cell engineering for tumor immunotherapy: From biology to clinical translation. Immunotherapy. 2012;4:703-18.
- [Google Scholar]
- The polarization of immune cells in the tumour environment by TGFbeta. Nat Rev Immunol. 2010;10:554-67.
- [Google Scholar]
- TGF-β signaling in myeloid cells is required for tumor metastasis. Cancer Discov. 2013;3:936-51.
- [Google Scholar]
- Guiding the “misguided” - Functional conditioning of dendritic cells for the DC-based immunotherapy against tumours. Eur J Immunol. 2011;41:18-25.
- [Google Scholar]
- Tumoral immune resistance mediated by enzymes that degrade tryptophan. Cancer Immunol Res. 2015;3:978-85.
- [Google Scholar]
- Lymph nodes and cancer metastasis: New perspectives on the role of intranodal lymphatic sinuses. Int J Mol Sci. 2016;18:E51.
- [Google Scholar]
- Induction of indoleamine 2,3-dioxygenase expression via heme oxygenase-1-dependant pathway during murine dendritic cell maturation. Biochem Pharmacol. 2010;80:491-505.
- [Google Scholar]
- PGE(2)-driven induction and maintenance of cancer-associated myeloid-derived suppressor cells. Immunol Invest. 2012;41:635-57.
- [Google Scholar]
- Wnt/β-catenin signaling in melanoma: Preclinical rationale and novel therapeutic insights. Cancer Treat Rev. 2016;49:1-2.
- [Google Scholar]
- TGF-β upregulates miR-181a expression to promote breast cancer metastasis. J Clin Invest. 2013;123:150-63.
- [Google Scholar]
- Engineered dendritic cells for gastrointestinal tumor immunotherapy: Opportunities in translational research. J Drug Target. 2013;21:126-36.
- [Google Scholar]
- The role of tumor initiating cells in drug resistance of breast cancer: Implications for future therapeutic approaches. Drug Resist Updat. 2010;13:99-108.
- [Google Scholar]
- Cancer stem-like cells derived from chemoresistant tumors have a unique capacity to prime tumorigenic myeloid cells. Cancer Res. 2014;74:2698-709.
- [Google Scholar]
- Interactions between cancer stem cells and their niche govern metastatic colonization. Nature. 2011;481:85-9.
- [Google Scholar]
- Exploring the role of cancer stem cells in radioresistance. Nat Rev Cancer. 2008;8:545-54.
- [Google Scholar]
- Comparative assessment of lipid based nano-carrier systems for dendritic cell based targeting of tumor re-initiating cells in gynecological cancers. Mol Immunol. 2016;79:98-112.
- [Google Scholar]
- Nanoengineered strategies to optimize dendritic cells for gastrointestinal tumor immunotherapy: From biology to translational medicine. Nanomedicine (Lond). 2014;9:2187-202.
- [Google Scholar]
- Comparative evaluation of hepatitis B surface antigen-loaded elastic liposomes and ethosomes for human dendritic cell uptake and immune response. Nanomedicine. 2010;6:110-8.
- [Google Scholar]
- Systemic and mucosal immune response induced by transcutaneous immunization using hepatitis B surface antigen-loaded modified liposomes. Eur J Pharm Sci. 2008;33:424-33.
- [Google Scholar]
- Polymerization of ethylene oxide, propylene oxide, and other alkylene oxides: Synthesis, novel polymer architectures, and bioconjugation. Chem Rev. 2016;116:2170-243.
- [Google Scholar]
- Type 1 and 2 immunity following vaccination is influenced by nanoparticle size: Formulation of a model vaccine for respiratory syncytial virus. Mol Pharm. 2007;4:73-84.
- [Google Scholar]
- Particle size and surface charge affect particle uptake by human dendritic cells in an in vitro model. Int J Pharm. 2005;298:315-22.
- [Google Scholar]
- Nanoparticles target distinct dendritic cell populations according to their size. Eur J Immunol. 2008;38:1404-13.
- [Google Scholar]
- Peripherally administered nanoparticles target monocytic myeloid cells, secondary lymphoid organs and tumors in mice. PLoS One. 2013;8:e61646.
- [Google Scholar]
- Administration routes affect the quality of immune responses: A cross-sectional evaluation of particulate antigen-delivery systems. J Control Release. 2010;147:342-9.
- [Google Scholar]
- Chitosan/pshRNA plasmid nanoparticles targeting MDR1 gene reverse paclitaxel resistance in ovarian cancer cells. J Huazhong Univ Sci Technolog Med Sci. 2009;29:239-42.
- [Google Scholar]
- Effect of cisplatin containing liposomes formulated by unsaturated chain-containing lipids on gynecological tumor cells. J Liposome Res. 2016;26:307-12.
- [Google Scholar]
- Development of synthetic of peptide-functionalized liposome for enhanced targeted ovarian carcinoma therapy. Int J Clin Exp Pathol. 2015;8:207-16.
- [Google Scholar]
- Subtype-specific binding peptides enhance the therapeutic efficacy of nanomedicine in the treatment of ovarian cancer. Cancer Lett. 2015;360:39-47.
- [Google Scholar]
- Multifunctional telodendrimer nanocarriers restore synergy of bortezomib and doxorubicin in ovarian cancer treatment. Cancer Res. 2017;77:3293-305.
- [Google Scholar]
- Administration of PLGA nanoparticles carrying shRNA against focal adhesion kinase and CD44 results in enhanced antitumor effects against ovarian cancer. Cancer Gene Ther. 2013;20:242-50.
- [Google Scholar]
- Improved therapeutic effect of folate-decorated PLGA-PEG nanoparticles for endometrial carcinoma. Bioorg Med Chem. 2011;19:4057-66.
- [Google Scholar]
- Metronomic docetaxel in PRINT nanoparticles and EZH2 silencing have synergistic antitumor effect in ovarian cancer. Mol Cancer Ther. 2014;13:1750-7.
- [Google Scholar]
- Reprogramming tumor-associated dendritic cells in vivo using miRNA mimetics triggers protective immunity against ovarian cancer. Cancer Res. 2012;72:1683-93.
- [Google Scholar]
- Engineering biodegradable guanidyl-decorated PEG-PCL nanoparticles as robust exogenous activators of DCs and antigen cross-presentation. Nanoscale. 2017;9:13413-8.
- [Google Scholar]
- Application of carbon nanoparticles in laparoscopic sentinel lymph node detection in patients with early-stage cervical cancer. PLoS One. 2017;12:e0183834.
- [Google Scholar]
- Telodendrimer nanocarrier for co-delivery of paclitaxel and cisplatin: A synergistic combination nanotherapy for ovarian cancer treatment. In: Biomaterials. Vol 37. 2015. p. :456-68.
- [Google Scholar]
- PEGylated dendritic diaminocyclohexyl-platinum (II) conjugates as pH-responsive drug delivery vehicles with enhanced tumor accumulation and antitumor efficacy. Biomaterials. 2014;35:10080-92.
- [Google Scholar]
- Toll-like receptor 3 agonist complexed with cationic liposome augments vaccine-elicited antitumor immunity by enhancing TLR3-IRF3 signaling and type I interferons in dendritic cells. Vaccine. 2012;30:4790-9.
- [Google Scholar]
- Surface-modified solid lipid nanoparticulate formulation for ifosfamide: Development and characterization. Nanomedicine (Lond). 2011;6:1397-412.
- [Google Scholar]
- Solid lipid nanoparticles for targeted brain drug delivery. Adv Drug Deliv Rev. 2007;59:454-77.
- [Google Scholar]
- Brain-targeted solid lipid nanoparticles containing riluzole: Preparation, characterization and biodistribution. Nanomedicine (Lond). 2010;5:25-32.
- [Google Scholar]
- STAT3 decoy oligodeoxynucleotides-loaded solid lipid nanoparticles induce cell death and inhibit invasion in ovarian cancer cells. PLoS One. 2015;10:e0124924.
- [Google Scholar]
- Hyaluronic acid decorated pluronic P85 solid lipid nanoparticles as a potential carrier to overcome multidrug resistance in cervical and breast cancer. Biomed Pharmacother. 2017;86:595-604.
- [Google Scholar]
- Design opportunities for actively targeted nanoparticle vaccines. Nanomedicine (Lond). 2008;3:343-55.
- [Google Scholar]
- Targeting nanoparticles to dendritic cells for immunotherapy. Methods Enzymol. 2012;509:143-63.
- [Google Scholar]
- Polyethylenimine-based micro/nanoparticles as vaccine adjuvants. Int J Nanomedicine. 2017;12:5443-60.
- [Google Scholar]
- Conditioning neoadjuvant therapies for improved immunotherapy of cancer. Biochem Pharmacol. 2017;145:12-7.
- [Google Scholar]
- Targeting with oligomannose-coated liposomes promotes maturation and splenic trafficking of dendritic cells in the peritoneal cavity. Int Immunopharmacol. 2011;11:164-71.
- [Google Scholar]
- Antigen co-encapsulated with adjuvants efficiently drive protective T cell immunity. Eur J Immunol. 2007;37:2063-74.
- [Google Scholar]
- Targeting cpG oligonucleotides to the lymph node by nanoparticles elicits efficient antitumoral immunity. J Immunol. 2008;181:2990-8.
- [Google Scholar]
- Targeted PLGA nano- but not microparticles specifically deliver antigen to human dendritic cells via DC-SIGN in vitro . J Control Release. 2010;144:118-26.
- [Google Scholar]
- In vivo targeting of dendritic cells in lymph nodes with poly(propylene sulfide) nanoparticles. J Control Release. 2006;112:26-34.
- [Google Scholar]
- LyP-1-conjugated nanoparticles for targeting drug delivery to lymphatic metastatic tumors. Int J Pharm. 2010;385:150-6.
- [Google Scholar]
- Targeting dendritic cells with antigen-containing liposomes: Antitumour immunity. Expert Opin Biol Ther. 2004;4:1735-47.
- [Google Scholar]
- Glycan-modified liposomes boost CD4+ and CD8+ T-cell responses by targeting DC-SIGN on dendritic cells. J Control Release. 2012;160:88-95.
- [Google Scholar]
- Targeting nanosystems to human DCs via fc receptor as an effective strategy to deliver antigen for immunotherapy. Mol Pharm. 2011;8:104-16.
- [Google Scholar]
- In vivo migration of dendritic cells labeled with synthetic superparamagnetic iron oxide. Int J Nanomedicine. 2011;6:2633-40.
- [Google Scholar]
- Monitoring of in vivo function of superparamagnetic iron oxide labelled murine dendritic cells during anti-tumour vaccination. PLoS One. 2011;6:e19662.
- [Google Scholar]
- Hypoxia-upregulated microRNA-630 targets dicer, leading to increased tumor progression. Oncogene. 2016;35:4312-20.
- [Google Scholar]
- Solid lipid nanoparticles carrying lipophilic derivatives of doxorubicin: Preparation, characterization, and in vitro cytotoxicity studies. J Microencapsul. 2016;33:381-90.
- [Google Scholar]
- Toll-like receptor 3-induced immune response by poly(d, l-lactide-co-glycolide) nanoparticles for dendritic cell-based cancer immunotherapy. Int J Nanomedicine. 2016;11:5729-42.
- [Google Scholar]
- Vaginal delivery of paclitaxel via nanoparticles with non-mucoadhesive surfaces suppresses cervical tumor growth. Adv Healthc Mater. 2014;3:1044-52.
- [Google Scholar]
- Paclitaxel and tacrolimus coencapsulated polymeric micelles that enhance the therapeutic effect of drug-resistant ovarian cancer. ACS Appl Mater Interfaces. 2016;8:4368-77.
- [Google Scholar]
- Folate receptor targeted delivery of siRNA and paclitaxel to ovarian cancer cells via folate conjugated triblock copolymer to overcome TLR4 driven chemotherapy resistance. Biomacromolecules. 2016;17:76-87.
- [Google Scholar]
- Photosensitizer enhanced disassembly of amphiphilic micelle for ROS-response targeted tumor therapy in vivo . Biomaterials. 2016;104:1-7.
- [Google Scholar]
- Raspberry-like poly(γ-glutamic acid) hydrogel particles for pH-dependent cell membrane passage and controlled cytosolic delivery of antitumor drugs. Int J Nanomedicine. 2016;11:5621-32.
- [Google Scholar]
- Effects of anti-CD44 monoclonal antibody IM7 carried with chitosan polylactic acid-coated nano-particles on the treatment of ovarian cancer. Oncol Lett. 2017;13:99-104.
- [Google Scholar]
- A pH-responsive glycolipid-like nanocarrier for optimising the time-dependent distribution of free chemical drugs in focal cells. Int J Pharm. 2017;522:210-21.
- [Google Scholar]
- Enhanced noscapine delivery using estrogen-receptor-targeted nanoparticles for breast cancer therapy. Anticancer Drugs. 2014;25:704-16.
- [Google Scholar]