Open access peer-reviewed chapter

Evolution of Organoids in Oncology

Written By

Allen Thayakumar Basanthakumar, Janitha Chandrasekhar Darlybai and Jyothsna Ganesh

Submitted: 11 January 2022 Reviewed: 04 March 2022 Published: 13 May 2022

DOI: 10.5772/intechopen.104251

From the Edited Volume

Organoid Bioengineering - Advances, Applications and Challenges

Edited by Manash K. Paul

Chapter metrics overview

201 Chapter Downloads

View Full Metrics

Abstract

An organoid is an in-vitro platform that recreates 3D multicellular aggerates to form tissues that fabricate the human cellular environment in the lab and imitate the functionalities of the specific organ or disease. Organoids effectively overcomes the gaps in research between 2D cell line and in-vivo models. For organoid development, both pluripotent stem cells and embryonic stem cells can be utilized, and recently Patient-Derived Organoids (PDO) was developed that overcome the limitations caused by using other cell lines. With the development of many advanced technologies in the field of research, the organoid evolution also progressed slowly into the development of patient-specific organ structures. Since tumor organoids were heterogeneous as well as patient-specific, it has many advantages that aid cancer therapy effectively. Apart from cancer treatment, organoids have a variety of applications in cancer research, the study of tissue-specific models, and also in the analysis of the relationship between tissue-specific cancer with various pathogens. Thus, the development of organoids in an effective way can pave the way for various biomedical applications. This chapter focuses on the trends in the journey of organoid research and the latest technologies developed specifically for organoids.

Keywords

  • organoid
  • 3D tissues
  • cancer research
  • cancer therapy

1. Introduction

Cancer is a heterogeneous disease that is caused by the progress of somatic mutations in normal cells [1]. Based on studies it was confirmed that continuous exposure to physical agents such as X-rays, gamma rays, UV rays, and genotoxic factors may end up in the progress of cancer cells from normal cells [2]. Carcinogenesis is a complex process that involves various pathways which need to be studied to understand the response and treatment method required for targeted therapy which is a complicated mechanism [3].

The development of drugs by using medicinal plants as bio source has also been studied extensively in various research [4]. It is essential to understand the biological interaction among immune cells among tumor immune microenvironment, stroma, and tumor for the success of cancer clinical treatments [5]. The overall cancer therapy may vary with multiple metastatic sites which in turn depend on the tumor heterogeneity [6]. The major biological complexities make the treatment methods difficult which in turn varies with the metastatic sites among individual patients [7]. The microenvironment of tumor cells includes normal stroma, malignant cells, and immune response based on every individual [8]. The durability and patient-to-patient response extremely varies based on each patient and hence very difficult to predict the consequences [9].

An organoid is an emerging technology with various applications in biomedical applications such as biobanking, disease modeling, regenerative medicine, and precision medicine [10]. An organoid is a technology used to fabricate 3D tissues in the laboratory that resembles parent tissue in function and structure and hence bridges the gap between 2D in-vitro and in-vivo models so that they can be utilized effectively in cancer research [11]. Though many cancer treatments were available the tumor heterogeneity limits the treatment as the drug sensitivity, drug invasion ability, growth rate of tumor changes based on individuals [12]. In the current review, the history and progressions in the developments of organoids were elaborated accompanied by their applications in cancer treatment along with their limits and steps required to overcome the limitations so that the organoid technology can be implemented efficiently in future research. Overview showed in Figure 1.

Figure 1.

Flowchart of use of normal organoids in making tumor organoids and utilization of tumor organoids in a biobank and its applications [13, 14, 15, 16, 17, 18].

Advertisement

2. The use of humanized models for cancer treatment

In the initial phase, the genetically engineered mouse models (GEMMs) provided a better appreciation for cancer treatment [19]. But later for practical applications, GEMMs technologies were identified to be expensive, laborious, and fetch up into complications when transformed into therapeutic applications [20]. One of the methods of generation of organoids is the utilization of Pluripotent stem cells (iPSCs), but the tissues generated from these embryonic stem cells were found to be phenotypically unstable and hence have the same limitations [21]. Later, Patient-derived xenografts (PDXs) were contemplated as a better replacement to conquer these constraints since, in this technology, models were provoked from an enormous pool of patients [22]. Unfortunately, these techniques also ended up in complications as the cancer cell lines generated in-vitro were genetically unstable and were devoid of the cellular microenvironment of tumor cells in-vivo and in most cases these cell lines ended up in unmatched cell lines from normal tissue which was considered as control cell lines [23].

Patient-derived tumor xenografts (PDTXs) is the recently available technology in cancer research that can maintain genomic stability and tumor heterogeneity but the major drawback of PDXs is that it is expensive and time-consuming and hence treatment gets delayed [13]. Later PDTXs technologies were upgraded by transplanting these cell lines into mouse models, but PDTXs failed to replicate the human-specific immune systems and were also ended up as a laborious and expensive process [14]. The clinical response of cancer treatment depends on the clinical model used for the study [24].

Advertisement

3. Organoids and their types

The unique way to improve cancer research has emerged with the help of organoid technology that accompanies tumor heterogeneity at low cost and with less time [25]. Cancer organoids serve as an effective tool to understand the interaction between tumor environment and genetic alterations [15]. The organoids derived from postnatal or adult tissue were termed ASC-derived organoids. In the case of ESCs and iPSCs derived organoids, the generation of organoids takes place from all three germ layers. For all these types of organoids, the growth of cells was carried out by using a series of differentiation protocols by utilizing growth factors and inhibitors in the process of organogenesis [11]. Recently many patient-derived organoids (PDTOs) have been developed that include liver, prostate cancer, and pancreatic cancer organoids [26]. A snapshot of types is shown in Figure 2.

Figure 2.

Development of 3D organoids culture [27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39].

To make more clarity to organoids, CRISPER gene-editing technology is being implemented to organoid to convert normal organoids into tumor organoids [25]. Upon various research, mutations have been induced in normal organoids like intestinal organoids [40], colon organoids [41], pancreatic organoids [42] to make them into tumor organoids that paves the way for flexible in-vitro cancer models. Apart from carcinogenesis, cancer organoids were also being implemented to study cancer metastasis which is the process of spreading cancer cells to other parts of the body [43]. Cancer organoids have also extended their applications in drug screening since PDTOs can be utilized to study gene expression, pathology, and tissue-specific genetic alterations [44]. Few researchers have used cancer organoids to generate tumor-reactive T cells that can be used in immunotherapy [45].

The brief details about various types of organoids have been discussed below:

3.1 Intestinal organoids

The pluripotent stem cells (iPSCs) were used to derive intestinal organoids that contain both mesenchymal and epithelial cells [27]. The basic intestinal crypt cells were allowed to grow in an appropriate medium containing a matrix that tends to organize into 3D epithelial cells that contain both physical and genetic resemblance of their parent organ [28].

3.2 Colon organoids

The colon organoids were originated from intestinal crypts in the presence of appropriate growth factors [29]. The stem cells of intestinal crypts differentiate into an epithelial complex that contains all types of intestinal cells. The isolated crypts develop into three-dimensional epithelial cells that form a sphere towards the lumen that further develops into colon organoids [30].

3.3 Pancreatic organoids

The capability of pancreatic ducts in in-vitro expansion and development of three-dimensional hollow structures in the presence of collagen as the suitable medium is utilized in the development of pancreatic organoids [31]. Many recent developments were carried out in establishing three-dimensional adult pancreatic organoids as well as fetal pancreatic organoids and both types of organoids were found to differ in morphology similar to that of parent cells [32].

3.4 Endometrial organoids

To investigate the biological processes involved in disease modeling, endometrial organoids were developed from primary endometrial cells of the human being [33]. For the development of three-dimensional endometrial organoids, the primary endometrial cells were dissociated, suspended in a Matrigel medium that promotes endometrial organ formation [34].

3.5 Lung organoids

The lung organoids were developed from stem cells of the lungs through a self-organization process [35, 36, 37]. The lung organoid technology has also been extended to develop various structures of lungs such as lung buds, airways, and alveolar cells that can be used to treat pulmonary diseases [38].

3.6 Ovarian organoids

Ovarian cancer is spread among a wide range of populations and the development of patient-specific ovarian organoids was found to be one of the precise approaches to enhance the treatment of ovarian cancer [39]. The ovarian organoid can enumerate the morphological heterogeneity of parent tissue and can aid in personalized therapy to enhance the treatment efficiency [46].

3.7 Breast organoids

Breast cancer is considered to be one of the most widely spread cancers among women with various subtypes and more heterogeneity [47]. Many studies have been performed on breast organoids and breast organoids can be grown either from epithelial cells [48] or from adult stem cells (ASCs) [27].

3.8 Retinal organoids

Retinal organoids were developed from Human Pluripotent Stem Cells (hPSCs) that is considered a labor-intensive and simple method of organoid development [49]. The retinal organoids also developed a physiological response to light to a certain extent that can be used in certain therapies [50].

3.9 Heart organoids

The heart organoids have proven to exhibit structural and functional features of developing human hearts [51]. The human Pluripotent Stem Cells (hPSCs) were used to develop in-vitro cardiac cell types that help in understanding and treatment of various cardiac diseases [52].

3.10 Brain organoids

The summons besotted in understanding the diseases caused in the central nervous system has been overcome by the development of brain organoids from either human Pluripotent Stem Cells (hPSCs) or epithelial cells [53].

Advertisement

4. Advantages of organoids over 2D techniques for cancer treatment

The development of organoid technology has overcome the limitations caused by 2D cell cultures in the following ways: 2D cultures were not repetitive of parent cultures while organoids were a portrait of their parent cells, the lack of predictivity faced in the use of 2D cultures was not visible while using organoid technology, and in 2D cell, culture models waste were being generated from growth medium which was overcome by organoid technology [54]. In organoid technology, multiple cell types were developed in-vivo that resemble the parent cells that have enormous applications in cancer research [11]. Tumor modeling is one of the vital applications of organoids as this technology help in identifying toxicity and effect of drugs before administering to patients which is not possible in 2D technology [40]. Organoids have strengthened the understanding of various disease progression and also helped scientists to study the pathways of disease carefully that enhance the treatment methods [55]. Organoids have extended their applications in various fields such as fundamental research [56], disease modeling [57], development of personalized medicine [58], and transplantation [59].

To overcome the technological limitations of cancer treatment, organoids were developed in which ASCs were proliferated in-vitro and self-organized into 3D structures by utilizing extracellular matrix proteins like BME and Matrigel as medium [60]. The main advantage of organoid technology is tissue-specific growth factors can be utilized based on the epithelial cells used for analysis [61]. Based on numerous studies, the ASCs derived organoids were found to be genetically and phenotypically stable and permit the expansion of stem cells in a required path which allows them to differentiate into tissue-specific cells [62]. Apart from tumor heterogeneity, more focus is required on lymphocyte infiltration trafficking along with the clinical response of individual patients to cancer treatment [63].

Advertisement

5. Stages of organoid development

Generally, the 3D culture of organoids was performed by using Matrigel derived from EHS (Engelbreth-Holam-Swarm) mouse sarcoma cells which were rich in adhesive proteins such as laminin, entactin, collagen, and proteoglycans. The Matrigel structural support, ECM signals along with the extracellular environment that supports the growth of cells [64]. In other techniques, Matrigel or fibroblasts were submerged in a medium to expose the upper layers of cells in the air. Later, the air-liquid interface will be utilized to culture the cells for better polarization and differentiation [65]. The organoid technology developed drastically after the successful generation of in-vitro organ generation from sponge cells [66], amphibians pronephros [67], chick embryos [68] followed by surface adhesion of cells using thermodynamic differentiation [69]. After the invention of pluripotent stem cells from mouse embryos, there was a significant impact on organoid development by utilizing stem cells [70].

The use of animal models has been considered in the initial days to predict the efficacy of drugs to tumor cells, later cell lines cultured in-vitro were used. But both these studies did not represent the actual model of tumor cells for which treatment is required [19]. A report by Radhakrishnan et al., 2017 [71] was used to analyze the immune target therapy for the treatment of cancer was carried out by utilizing the CANscript exvivo platform technology.

The various stages of organoid development were explained below [72] and the overview showed in Figure 3.

Figure 3.

Flowchart of various stages of organoid development [10, 19].

  • 1900–1950: In the early 1900s hanging drop culture method was employed in organ development followed by tube cultures. Subsequently in 1907 organoids were developed from sponge cells and later in 1944 organoid development was carried out from amphibian pronephros.

  • 1950–1980: In 1952 organoids were developed from chick embryos. In 1975 floating collagen cells were developed followed by characterization of laminin using organoid technology in 1979.

  • 1980–2000: The isolation of pluripotent stem cells from mouse tails was successfully performed in 1981 which paved the way for great progress in organoid technology in subsequent years. The development of human blastocytes was performed in 1998.

  • 2000–2020: The development of the first organoid from pluripotent stem cells was performed in 2006 from mouse fibroblast, followed by the development of organoids from cortex tissue, adult stem cells, development of gastric organoids, development of retinal, kidney, pancreas, lungs, fallopian tubes and snake venom were performed successfully in subsequent years.

Advertisement

6. Role of organoids in drug development and cancer research

Apart from therapeutic applications, organoids can also be employed in cancer research to study the mechanism of cancer progression and the interaction between pathogens and organs in the development of cancer cells. In a study conducted by Scanu et al., 2016 [73] the interaction of pathogen Salmonella enterica with the gall bladder cells was found to provoke gall bladder carcinoma. Similarly, in a study conducted by Yin et al., 2015 [74] relations between hepatic virus and liver cell carcinoma have been studied extensively.

Organoid technology has also been implemented to study the genetic interaction between mutation and progression of various types of cancer [75]. Organoids guide in the study of the initiation of tumors with the progression of cancer at various genetic levels [76].

Organoids can be used to study the response of drugs to each patient so can be utilized in drug development for cancer research [77]. Though many drugs were working on cancer models they get eliminated in final clinical trials due to unbearable side effects or lack of efficiency compared to clinical models [78]. Many biobanks of organoids were used to study the efficiency of novel drugs [16]. By utilizing organoids in drug screening, it will be feasible to predict the patient’s response and value of Patient-Derived Organoids in chemotherapy. The data obtained from PDOs can be used to predict the outcome of individual patients which was not possible by using the cell line technique [17].

Advertisement

7. The use of biobanks for organoid research

The progression on the development of organoids from ASCs paved the way for the establishment of the biobank for cancer [18]. The cancer biobanks provide cancer organoid cultures of numerous cancer subtypes [79]. Based on an investigation of numerous organoids using biobanks it was concluded that the dependency of organoids on ligands varies from one type of organoids to other types of organoids [59]. The inception of biobanks of organoids for cancer paved the way for the up-gradation of anti-cancer drugs with a wide range of testing based on requirements by cancer patients [21]. In a study done by Smalley and coworkers in 2018 [80], CD 34 biomarker has been utilized effectively in cancer treatment and the outcome has been studied in detail.

Advertisement

8. Technological developments of organoids

The existing in-vitro and in-vivo platforms used for cancer therapy were proved to be not much effective and hence there is a need to derive many new platforms for cancer treatment [81]. In a study conducted by Majumder et al., 2015 [82], an attempt has been carried out to overcome the tumor heterogeneity with the help of the CANScriptTM platform designed by their team. In 2013, Radhakrishnana and his coworkers [83] attempted to design a drug PAT-1102 that acts as a HDAC (Histone deacetylase) inhibitor and hence can be utilized for cancer treatment. The utilization of Light-screening fluorescence microscopy, confocal microscopy, and multiphoton laser screening microscopy in organoid development can be used to visualize the 3D image of polarized and nonpolarized cells based on the individual organoid model [84]. Organoids have been utilized effectively to understand human brain development and also about various diseases elaborately [85]. Apart from cancer research organoids were also used to study various infectious diseases [86], genetic diseases by combing with CRISPER technology [87], to study the gene function [88] and cell development [89].

The development of organoids paved the way for 3D imaging technology which can be used to visualize complex organizations that were not possible using 2D imaging technology [90]. The 3D imaging technology helped scientists to visualize cellular components, intracellular processes, and architecture of cells in a detailed manner [91]. Lungs were considered as one of the most complex organs in the human body with numerous types of cells and the development of lung organoids had a significant impact on the treatment of lung cancer, asthma, cystic fibrosis, and pulmonary fibrosis [92]. Based on 3D imaging technology major therapeutic drugs were developed that can be used successfully in lung cancer treatment [93]. The 3D models increased the predictability and reliability of preclinical assays and decreased the use of animal models [94]. The quantitative analysis of the organization of various cell types in aggregates uses 3D imaging technology for detailed analysis [95].

Another technological up-gradation seen in organoid technology is Organs-on-a-chip which is used to model various functional groups of organs for detailed study [96]. Initially, the anatomy of the particular organ was studied, and then its basic elements were investigated that can be used to study organ-specific physical and biochemical applications [97]. Retinal diseases were one of the major causes of vision loss in humans worldwide while the complexity of neuro-retinal organization and complex blood supply causes side effects by the use of therapeutic drugs in the treatment of retinal diseases [98]. The development of retinal organoids improved the treatment methods of retinal diseases and also organs-on-a-chip technology paved the way to study cell types in detail that can further enhance the treatment methodology of retinal disease [99]. In case the focus of cancer research is based on a particular cell type or organ miniature types of organoids can be developed by utilizing organ-on-a-chip technology [100]. The organoid development and organoid-on-a-chip technology has their unique characteristics and limitations and based on research requirement the suitable technology is to be utilized [101]. The usage of organoid-on-a-chip technology on brain organoids paved the way to cure many neurological diseases [102].

In earlier days, tumor spheroids were developed to analyze the capability of antitumor therapeutic drugs, and recently tissue-specific organoids were utilized effectively to model various organs that can be used in cancer treatment by overcoming the limitations of ethical concerns caused by tumor spheroids [103]. The use of brain organoids to study neural diseases has proven that the brain organoids developed in 3D cultures express a large number of genes entangled in neurological problems and hence make the study process feasible compared to 2D technology [104]. In recent areas of research organoid technology is being implemented extensively in image-based phenotypic high throughput screening [105].

Advertisement

9. Limitations of organoids

Generation of organoids from epithelial cells may lead to contamination or overgrowth of normal cells [58]. Generally, the organoids developed from cancer cells will have less growth than organoids developed from normal cells which result in overgrowth of normal cells in the medium which can be overcome by limiting the use of growth factors in the medium [106]. Upon analyzing the efficiency of various organoids on drug development in-vitro, a positive predictive value of 88% and a negative predictive value of 100% were endorsed upon numerous studies [107, 108]. The organoids developed in-vitro lack the native microenvironment like stromal cells, immune cells [109]. This limitation can be overcome by culturing the tissues with required cellular elements for proper differentiation but this may end up in more cost [110]. In a study by [111], a high-throughput screening accompanied with patient-derived 3D organoids has been successfully utilized to overcome the limitations of organoids in drug screening. In another study, an organ-on-a-chip platform has been devised which contains hollow microchannels filled with living cells resembling human organs and organoids which can be utilized effectively for drug screening [112]. Another major limitation in organoid development is lack of reproducibility, laborious and costly method [113].

Advertisement

10. Conclusions

Cancer is a heterogeneous disease caused by a mutation in normal cells that results in the abnormal growth of cells. Though many treatment methods and drugs were available in cancer treatment, there is no proper cure, and the drugs used ended up in major side effects in patients. The development of cancer cells varies with the patient and also based on the type of cancer. Thus, there was a need to derive a patient-specific treatment method is cancer treatment. But the limitation of these 2D cell line models where it was a time-consuming process, expensive, and sometimes cannot meet the requirement of tumor heterogeneity to large extent. To overcome this many technologies such as GEMMs and PDTXs were developed and they have been proved to be effective techniques. To overcome these limitations organoid technology has emerged in which tissues were grown in in-vitro from cells derived from patients and these tissues can be utilized for drug development. Apart from drug development in cancer research organoid technology also paved the way to study elaborately the mechanisms behind cancer treatment, about particular organs in detail, and also the relationship between pathogens and various types of cancer. The culturing of organoids along with immune cells has shown successive progress in creating better models and understanding the cancer progression in many research works. Though organoid technology also has various limitations various methods have been established to overcome the limitations and hence organoids can be used as an effective tool in cancer research and cancer treatment.

Acknowledgments

We would like to thank Ms. Amudha Madhavan for her critical review and proof reading of the chapter.

Acronyms and abbreviations

GEMMs

Genetically Engineered Mouse Models

PDTXs

Patient-derived xenografts

PDTOs

patient-derived organoids

iPSCs

pluripotent stem cells

human Pluripotent Stem Cells

hPSCs

Adult Stem Cells

ASCs

References

  1. 1. Stratton MR, Campbell PJ, Futreal PA. The cancer genome. Nature. 2009;458(7239):719-724
  2. 2. Luch A. Nature and nurture-lessons from chemical carcinogenesis. Nature Reviews. Cancer. 2005;5(2):113-125
  3. 3. Radhakrishnan P, Baraneedharan U, Veluchamy S, Dhandapani M, Pinto DD, Thiyagarajan S, et al. Inhibition of rapamycin-induced AKT activation elicits differential antitumor response in head and neck cancers. Cancer Research. 2013;73(3 :1118-1127
  4. 4. Starlin T, Saravana Prabha P, Allen Thayakumar B, Gopalakrishnan VK. Screening and GC-MS profiling of ethanolic extract of Tylophora pauciflora. Bioinformation. 2019;15(6):425-429
  5. 5. Smalley M, Natarajan S, Mondal J, Best D, Goldman D, Shanthappa B, Pellowe M, et al. Nanoengineered disruption of heat shock protein 90 targets drug-induced resistance and relieves natural killer cell suppression in breast cancer. Cancer Research. 2020;80(23):5355-5366
  6. 6. Ikpeazu C, Smalley M, Ulaganathan B, Thayakumar A, Majeiko L, Ganesh J, et al. Abstract LB-346: Case study: Non-uniform response to therapy in multiple metastatic is predicted using CANscript, a live tissue, ex-vivo, platform. AACR Annual Meeting. 2018;78:(13). DOI: 10.1158/1538-7445.AM2018-LB-346
  7. 7. Ikpeazua C, Smalley M, Ulaganathan B, Thayakumar A, Basavaraja S, Gertje H, et al. Profiling metastatic lesions from a pembro-refractory patient to reveal distinct genomic instabilities and non-uniform response to drug combinations, ex vivo. Journal of Clinical Oncology. 2018;36:15
  8. 8. Smalley M, Shanthappa BU, Gertje H, Lawson M, Ulaganathan B, Thayakumar A, et al. Therapy-induced priming of natural killer cells predicts patient-specific tumor rejection in multiple breast cancer indications. SABCS. 2017;630
  9. 9. Smalleyc M, Shanthappa BU, Gertje H, Lawson M, Ulaganathan B, Thayakumar A, et al. Characterizing immunotherapy-induced lymphocyte infiltration at the single patient level using CANscriptTM, an ex-vivo human tumor model. Immunotherapy of cancer. 2017;5
  10. 10. Corro C, Novellasdemunt L, Li XVSW. A brief history of organoids. American Journal of Physiology. Cell Physiology. 2020;319:C151-C165
  11. 11. Drost J, Clevers H. Organoids in cancer research. Nature Reviews. Cancer. 2018;18(7):407-418
  12. 12. McGranahan N, Swanton C. Clonal heterogeneity and tumor evolution: Past, present, and the future. Cell. 2017;168(4):613-628
  13. 13. Bleijs M, van deWeteringM, Clevers H, Drost J. Xenograft and organoid model systems in cancer research. The EMBO Journal. 2019;38(15):e101654
  14. 14. Sundaram M, Ulaganathan B, Dhandapani M, Thayakumar A, Mukhopadhyay P, Thiyagarajan S, et al. Abstract 3736: Application of patient tumors-derived tumor ecosystem platform for the development of novel HDAC inhibitor in solid cancers. AACR Annual Meeting. 2014;74
  15. 15. Fan H, Demirci U, Chen P. Emerging organoid models: Leaping forward in cancer research. Journal of Hematology & Oncology. 2019;12:142
  16. 16. Sachs N, de Ligt J, Kopper O, Gogola E, Bounova G, Weeber F, et al. A living biobank of breast cancer organoids captures disease heterogeneity. Cell. 2018;172(373-86):e10
  17. 17. Boretto M, Maenhoudt N, Luo X, Hennes A, Boeckx B, Bui B, et al. Patient-derived organoids from endometrial disease capture clinical heterogeneity and are amenable to drug screening. Nature Cell Biology. 2019;21:1041-1051
  18. 18. Driehuis E, Kretzschmar K, Clevers H. Establishment of patient-derived cancer organoids for drug screening applications. Nature Protocols. 2020;15(10):3380-3409
  19. 19. Majumder B, Baraneedharan U, Thiyagarajan S, Radhakrishnan P, Narasimhan H, Dhandapani M, et al. Predicting clinical response to anticancer drugs using an ex vivo platform that captures tumour heterogeneity. Nature Communications. 2015;6:6169
  20. 20. Holloway EM, Capeling MM, Spence JR. Biologically inspired approaches to enhance human organoid complexity. Development. 2019;146(8):dev166173
  21. 21. Junttila MR, de Sauvage FJ. Influence of tumour microenvironment heterogeneity on therapeutic response. Nature. 2013;501:346-354
  22. 22. Sachs N, Clevers H. Organoid cultures for the analysis of cancer phenotypes. Current Opinion in Genetics & Development. 2014;24:68-73
  23. 23. Shroyer NF. Tumor organoids fill the niche. Cell Stem Cell. 2016;18(6):686-687
  24. 24. Fatehullah A, Tan SH, Barker N. Organoids as an in vitro model of human development and disease. Nature Cell Biology. 2016;18(3):246
  25. 25. Onuma K, Ochiai M, Orihashi K, Takahashi M, Imai T, Nakagama H, et al. Genetic reconstitution of tumorigenesis in primary intestinal cells. Proceedings of the National Academy of Sciences. 2013;110(27):11127-11132
  26. 26. Broutier L, Mastrogiovanni G, Verstegen MM, Francies HE, Gavarro LM, Bradshaw CR, et al. Human primary liver cancer–derived organoid cultures for disease modeling and drug screening. Nature Medicine. 2017;23(12):1424
  27. 27. Sato T, Clevers H. SnapShot: Growing organoids from stem cells. Cell. 2015;161:1700-1700.e1
  28. 28. Sato T, Stange DE, Ferrante M, Vries RG, Van Es JH, Van den Brink S, et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett's epithelium. Gastroenterology. 2011;141:1762-1772
  29. 29. Sato T, Clevers H. Growing self-organizing mini-guts from a single intestinal stem cell: Mechanism and applications. Science. 2013;340:1190-1194
  30. 30. Sebert M, Denadai-Souza A, Quaranta M, Racaud-Sultan C, Chabot S, Lluel P, et al. Thrombin modifies growth, proliferation and apoptosis of human colon organoids: A protease-activated receptor 1- and protease-activated receptor 4-dependent mechanism. British Journal of Pharmacology. 2018;175:3656-3668
  31. 31. Kerr-Conte J, Pattou F, Lecomte-Houcke M, Xia Y, Boilly B, Proye C, et al. Ductal cyst formation in collagen-embedded adult human islet preparations. A means to the reproduction of nesidioblastosis in vitro. Diabetes. 1996;45(8):1108-1114
  32. 32. Lee J, Sugiyama T, Liu Y, Wang J, Gu X, Lei J, et al. Expansion and conversion of human pancreatic ductal cells into insulin-secreting endocrine cells. eLife. 2013;2:e00940
  33. 33. Turco MY, Gardner L, Hughes J, Cindrova-Davies T, Gomez MJ, Farrell L, et al. Long-term, hormone-responsive organoid cultures of human endometrium in a chemically defined medium. Nature Cell Biology. 2017;19:568
  34. 34. Boretto M, Cox B, Noben M, Hendriks N, Fassbender A, Roose H, et al. Development of organoids from mouse and human endometrium showing endometrial epithelium physiology and long-term expandability. Development. 2017;144:1775-1786
  35. 35. Lancaster MA, Knoblich JA. Organogenesis in a dish: Modeling development and disease using organoid technologies. Science. 2014;345(6194):1247125
  36. 36. Lee JH, Bhang DH, Beede A, Huang TL, Stripp BR, Bloch KD, et al. Lung stem cell differentiation in mice directed by endothelial cells via a BMP4-NFATc1-thrombospondin-1 axis. Cell. 2014;156(3):440-455
  37. 37. Wilkinson DC, Alva-Ornelas JA, Sucre JMS, Vijayaraj P, Durra A, Richardson W, et al. Development of a three-dimensional bioengineering technology to generate lung tissue for personalized disease modeling. Stem Cells Translational Medicine. 2017;6(2):622-633
  38. 38. McCauley KB, Hawkins F, Serra M, Thomas DC, Jacob A, Kotton DN. Efficient derivation of functional human airway epithelium from pluripotent stem cells via temporal regulation of Wnt signaling. Cell Stem Cell. 2017;20(6): 844-57. e6
  39. 39. Koshiyama M, Matsumura N, Konishi I. Recent concepts of ovarian carcinogenesis: Type I and type II. BioMed Research International. 2014;934261
  40. 40. Matano M, Date S, Shimokawa M, Takano A, Fujii M, Ohta Y, et al. Modeling colorectal cancer using CRISPR-Cas9–mediated engineering of human intestinal organoids. Nature Medicine. 2015;21(3):256
  41. 41. Nakayama M, Sakai E, Echizen K, Yamada Y, Oshima H, Han T, et al. Intestinal cancer progression by mutant p53 through the acquisition of invasiveness associated with complex glandular formation. Oncogene. 2017;36(42):5885
  42. 42. Schell MJ, Yang M, Teer JK, Lo FY, Madan A, Coppola D, et al. A multigene mutation classification of 468 colorectal cancers reveals a prognostic role for APC. Nature Communications. 2016;7:11743
  43. 43. Gomez-Cuadrado L, Tracey N, Ma R, Qian B, Brunton VG. Mouse models of metastasis: Progress and prospects. Disease Models & Mechanisms. 2017;10(9):1061-1074
  44. 44. Sachs N, de Ligt J, Kopper O, Gogola E, Bounova G, Weeber F, et al. A living biobank of breast cancer organoids captures disease heterogeneity. Cell. 2018;172(1-2):373-386.e10
  45. 45. Della Corte CM, Barra G, Ciaramella V, Di Liello R, Vicidomini G, Zappavigna S, et al. Antitumor activity of dual blockade of PD-L1 and MEK in NSCLC patients derived three-dimensional spheroid cultures. Journal of Experimental & Clinical Cancer Research. 2019;38(1):253
  46. 46. Kopper O, de Witte CJ, Lohmussaar K, Valle-Inclan JE, Hami N, Kester L, et al. An organoid platform for ovarian cancer captures intra- and interpatient heterogeneity. Nature Medicine. 2019;25:838-849
  47. 47. Lakhani SR. WHO Classification of Tumours of the Breast. 4th ed. Lyon: International Agency for Research on Cancer; 2012
  48. 48. Lee GY, Kenny PA, Lee EH, Bissell MJ. Three-dimensional culture models of normal and malignant breast epithelial cells. Nature Methods. 2007;4:359-365
  49. 49. Vining KH, Mooney DJ. Mechanical forces direct stem cell behavior in development and regeneration. Nature Reviews. Molecular Cell Biology. 2017;18:728-742
  50. 50. Zhong X, Gutierrez C, Xue T, Hampton C, Vergara MN, Cao LH, et al. Generation of three-dimensional retinal tissue with functional photoreceptors from human iPSCs. Nature Communications. 2014;2014(5):4047
  51. 51. Zhang Q , Schepis A, Huang H, Yang J, Ma W, Torra J, et al. Designing a green fluorogenic protease reporter by flipping a beta strand of GFP for imaging apoptosis in animals. Journal of the American Chemical Society. 2019;141:4526-4530
  52. 52. Dunn KK, Palecek SP. Engineering scalable manufacturing of high-quality stem cell-derived cardiomyocytes for cardiac tissue repair. Frontiers in Medicine. 2018;5:1-18
  53. 53. Adams JW, Cugola FR, Muotr AR. Brain organoids as tools for modeling human neurodevelopmental disorders. Physiology. 2019;34:365-375
  54. 54. Ho BX, Qian Pek NM, Soh B-S. Disease modeling using 3D organoids derived from human induced pluripotent stem cells. International Journal of Molecular Sciences. 2018;19:936
  55. 55. Nantasanti S, de Bruin A, Rothuizen J, Penning LC, Schotanus BA. Concise review: Organoids are a powerful tool for the study of liver disease and personalized treatment Design in Humans and Animals. Stem Cells Translational Medicine. 2016;5(3):325-330
  56. 56. Zhou J, Su J, Fu X, Zheng L, Yin Z. Microfluidic device for primary tumor spheroid isolation. Experimental Hematology & Oncology. 2017;6:22
  57. 57. Ben-David U, Ha G, Tseng YY, Greenwald NF, Oh C, Shih J, et al. Patient-derived xenografts undergo mouse-specific tumor evolution. Nature Genetics. 2017;49:1567-1575. DOI: 10.1038/ng.3967
  58. 58. Vlachogiannis G, Hedayat S, Vatsiou A, Jamin Y, Fernandez-Mateos J, Khan K, et al. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science. 2018;359:920-926
  59. 59. Seino T, Kawasaki S, Shimokawa M, Tamagawa H, Toshimitsu K, Fujii M, et al. Human pancreatic tumor organoids reveal loss of stem cell niche factor dependence during disease progression. Cell Stem Cell. 2018;22(3):454-67. e6
  60. 60. Sato T, Vries RG, Snippert HJ, van de Wetering M, Barker N, Stange DE, et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature. 2009;459(7244):262-265
  61. 61. Ootani A, Li X, Sangiorgi E, Ho QT, Ueno H, Toda S, et al. Sustained in vitro intestinal epithelial culture within a Wnt dependent stem cell niche. Nature Medicine. 2009;15(6):701-706
  62. 62. Serra D, Mayr U, Boni A, Lukonin I, Rempfler M, Challet Meylan L, et al. Self-organization and symmetry breaking in intestinal organoid development. Nature. 2019;569(7754):66-72
  63. 63. Smalleyd M, Shanthappa B, Gertje H, Lawson M, Ulaganathan B, Thayakumar A, et al. Nonuniform T-cell infiltration induced by PD-1 checkpoint blockade, ex vivo, predicts distinct clinical response. Cancer Research. 2018;13:78
  64. 64. Orkin RW, Gehron P, McGoodwin EB, Martin GR, Valentine T, Swarm RA. Murine tumor producing a matrix of basement membrane. The Journal of Experimental Medicine. 1977;145:204-220
  65. 65. Turner DA, Baillie-Johnson P, Martinez AA. Organoids and the genetically encoded self-assembly of embryonic stem cells. BioEssays. 2016;38:181-191
  66. 66. Wilson HV. A new method by which sponges may be artificially reared. Science. 1907;25:912-915
  67. 67. Holtfreter J. Experimental studies on the development of the pronephros. Revue Canadienne de Biologie. 1944;3:220-250
  68. 68. Weiss P, Taylor AC. Reconstitution of complete organs from single-cell suspensions of chick embryos in advanced stages of differentiation. Proceedings National Academic Science USA. 1960;46:1177-1185
  69. 69. Steinberg MS. The problem of adhesive selectivity in cellular interactions. In: Locke M, editor. Cellular Membranes in Development. New York and London: Academic Press; 1964
  70. 70. Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian S, et al. Induced pluripotent stem cell lines derived from human somatic cells. Science. 2007;318:1917-1920
  71. 71. Radhakrishnan P, Goldman A, Ulaganathan B, Kumar AT, Maciejko L, Gertje H, et al. Predicting tumor-immune response to checkpoint inhibitors using a novel patient-derived live tumor explant model. Journal of Clinical Oncology. 2017;35(15_suppl):e20035-e20035
  72. 72. Simian M, Bissell MJ. Organoids: A historical perspective of thinking in three dimensions. The Journal of Cell Biology. 2017;216(1):31-40
  73. 73. Scanu T, Spaapen RM, Bakker JM, Pratap CB, Wu LE, Hofland I, et al. Salmonella manipulation of host signaling pathways provokes cellular transformation associated with gallbladder carcinoma. Cell Host & Microbe. 2015;17:763-774
  74. 74. Yin Y, Bijvelds M, Dang W, Xu L, van der Eijk AA, Knipping K, et al. Modeling rotavirus infection and antiviral therapy using primary intestinal organoids. Antiviral Research. 2015;123:120-131
  75. 75. Roerink SF, Sasaki N, Lee-Six H, Young MD, Alexandrov LB, Behjati S, et al. Intra-tumour diversification in colorectal cancer at the single-cell level. Nature. 2018;556:457-462
  76. 76. Xu H, Lyu X, Yi M, Zhao W, Song Y, Wu K. Organoid technology and applications in cancer research. Journal of Hematology & Oncology. 2018;11:116
  77. 77. Abbasi J. Patient-derived organoids predict cancer treatment response. Journal of the American Medical Association. 2018;319:1427
  78. 78. Viardot A, Bargou R. Bispecific antibodies in haematological malignancies. Cancer Treatment Reviews. 2018;65:87-95
  79. 79. Jacob F, Salinas RD, Zhang DY, Nguyen PTT, Schnoll JG, Wong SZH, et al. A patient derived glioblastoma organoid model and biobank recapitulates inter- and intratumoral heterogeneity. Cell. 2020;180(1):188-204
  80. 80. Smalleye M, Alam N, Murmu N, Somashekhar SP, Ulaganathan B, Thayakumar A, et al. A live tissue platform allows dynamic measurement of neovascularization and prediction of clinical response in human breast cancer samples, ex vivo. SABCS. 2018
  81. 81. Sundarama M, Thiyagarajan S, Dhandapani M, Brijwani N, Prasath A, Thayakumar A, et al. Human tumor derived Oncoprint® platform predicts molecular mechanism of sensitivity and resistance to Fragmin in pancreatic cancer. Cancer Research. 2013;8:73
  82. 82. Majumder B, Ulaganathan B, Thayakumar A, Thiyagarajan S, Brijiwani N, Tewari B, et al. Abstract 1304: Identification of responders for Anti-CTLA4 in refractory colorectal cancers using CANScript™ platform. AACR 106th Annual Meeting. 2015;75
  83. 83. Radhakrishnana P, Dhandapani M, Ulaganathan B, Thayakumar A, Thiyagarajan S, Pinto D, et al. Majumde P K. PAT-1102, a novel HDAC inhibitor exhibits potent anti-tumor efficacy in patient-derived refractory solid tumors. Cancer Research. 2013;73:8
  84. 84. Rios AC, Clevers H. Imaging organoids: A bright future ahead. Nature Methods. 2018;15(1):24-26
  85. 85. Kit-Yi Yam A. 3D Perspective on Human Organoids: Promising Models for Studying Brain Development and Disease. Life canvas technologies; 2020
  86. 86. Garcez PP, Loiola EC, Madeiro da Costa R, Higa LM, Trindade P, Delvecchio R, et al. Zika virus impairs growth in human neurospheres and brain organoids. Science. 2016;352:816-818
  87. 87. Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science. 2012;337:816-821
  88. 88. Tetteh PW, Basak O, Farin HF, Wiebrands K, Kretzschmar K, Begthel H, et al. Replacement of lost Lgr5-positive stem cells through plasticity of their enterocyte-lineage daughters. Cell Stem Cell. 2016;18:203-213
  89. 89. Basak O, Beumer J, Wiebrands K, Seno H, van Oudenaarden A, Clevers H. Induced quiescence of Lgr5+ stem cells in intestinal organoids enables differentiation of hormone-producing enteroendocrine cells. Cell Stem Cell. 2017;20:177-190 e4
  90. 90. Richardson DS, Lichtman JW. SnapShot: Tissue clearing. Cell. 2017;171:496-496.e1
  91. 91. Jamieson PR, Dekkers JF, Rios AC, Fu NY, Lindeman GJ amd Visvader JE. Derivation of a robust mouse mammary organoid system for studying tissue dynamics. Development. 2017;144(6):1065-1071
  92. 92. Kim M, Mun H, Sung CO, Cho EJ, Jeon HJ, Chun SM, et al. Patient-derived lung cancer organoids as in vitro cancer models for therapeutic screening. Nature Communications. 2019;10(1):3991
  93. 93. Barkauskas CE, Chung MI, Fioret B, Gao X, Katsura H, Hogan BL. Lung organoids: Current uses and future promise. Development. 2017;144(6):986-997
  94. 94. Breslin S, O'Driscoll L. Three-dimensional cell culture: The missing link in drug discovery. Drug Discovery Today. 2013;18:240-249
  95. 95. Pampaloni F, Reynaud EG, Stelzer EHK. The third dimension bridges the gap between cell culture and live tissue. Nature Reviews. Molecular Cell Biology. 2007;8:839-845
  96. 96. Huh D, Hamilton GA, Ingber DE. From 3D cell culture to organs-on-chips. Trends in Cell Biology. 2011;21:745-754
  97. 97. Yum K, Hong SG, Healy KE, Lee LP. Physiologically relevant organs on chips. Biotechnology Journal. 2014;9:16-27
  98. 98. Renouf DJ, Velazquez-Martin JP, Simpson R, Siu LL, Bedard PL. Ocular toxicity of targeted therapies. Journal of Clinical Oncology. 2012;30:3277-3286
  99. 99. Zhong X, Gutierrez C, Xue T, et al. Generation of three-dimensional retinal tissue with functional photoreceptors from human iPSCs. Nature Communications. 2014;5:4047
  100. 100. Dutta D, Heo I, Clevers H. Disease modeling in stem cell-derived 3D organoid systems. Trends in Molecular Medicine. 2017;23:393
  101. 101. Bhatia SN, Ingber DE. Microfluidic organs-on-chips. Nature Biotechnology. 2014;32:760
  102. 102. Wang Y, Wang L, Zhu Y, Qin J. Human brain organoid-on-a-chip to model prenatal nicotine exposure. Lab on a Chip. 2018;18:851-860
  103. 103. Munsie M, Hyun I, Sugarman J. Ethical issues in human organoid and gastruloid research. Development. 2017;144:942-945
  104. 104. Tekin H, Simmons S, Cummings B, Gao L, Adiconis X, Hession C. C, Ghoshal A, Dionne D, Choudhury S. R, Yesilyurt V et al. Effects of 3D culturing conditions on the transcriptomic profile of stem-cell-derived neurons. Nature Biomedical Engineering. 2018:1-15
  105. 105. Lukonin I, Zinner M, Liberali P. Organoids in image-based phenotypic chemical screens. Experimental & Molecular Medicine. 2021;53:1495-1502
  106. 106. Drost J, van Jaarsveld RH, Ponsioen B, Zimberlin C, van Boxtel R, Buijs A, et al. Sequential cancer mutations in cultured human intestinal stem cells. Nature. 2015;521(7550):43-47
  107. 107. Yao Y, Xu X, Yang L, Zhu J, Wan J, Shen L, et al. Patient-derived organoids predict chemoradiation responses of locally advanced rectal cancer. Cell Stem Cell. 2020;26(1):17-26
  108. 108. Ooft SN, Weeber F, Dijkstra K, McLean CM, Kaing S, Vanwerkhoven E, et al. Patient-derived organoids can predict response to chemotherapy in metastatic colorectal cancer patients. Science Translational Medicine. 2019;11(513):eaay2574
  109. 109. Jabs J, Zickgraf FM, Park J, Wagner S, Jiang X, Jechow K, et al. Screening drug effects in patient-derived cancer cells links organoid responses to genome alterations. Molecular Systems Biology. 2017;13:955
  110. 110. Workman MJ, Mahe MM, Trisno S, Poling HM, Watson CL, Sundaram N, et al. Engineered human pluripotent-stem-cell-derived intestinal tissues with a functional enteric nervous system. Nature Medicine. 2017;23:49-59
  111. 111. Boussaad I, Cruciani G, Bolognin S, Antony P, Dording CM, Kwon YJ, et al. Integrated, automated maintenance, expansion and differentiation of 2D and 3D patient-derived cellular models for high throughput drug screening. Scientific Reports. 2021;11:439
  112. 112. Kasendra M, Tovaglieri A, Sontheimer-Phelps A, Jalili-Firoozinezhad S, Bein A, Chalkiadaki A, et al. Development of a primary human small intestine-on-a-Chip using biopsy-derived organoids. Scientific Reports. 2018;8:287
  113. 113. Dayem AA, Lee SB, Kim K, Lim KM, Jeon TI, Cho SG. Recent advances in organoid culture for insulin production and diabetes therapy: Methods and challenges. BMB Reports. 2019;52(5):295-303

Written By

Allen Thayakumar Basanthakumar, Janitha Chandrasekhar Darlybai and Jyothsna Ganesh

Submitted: 11 January 2022 Reviewed: 04 March 2022 Published: 13 May 2022