Next Article in Journal
New Record of Dendronephthya sp. (Family: Nephtheidae) from Mediterranean Israel: Evidence for Tropicalization?
Next Article in Special Issue
The Capacity of Magnesium to Induce Osteoclast Differentiation Is Greatly Enhanced by the Presence of Zoledronate
Previous Article in Journal
Identification of a Novel Bioactive Peptide Derived from Frozen Chicken Breast Hydrolysate and the Utilization of Hydrolysates as Biopreservatives
Previous Article in Special Issue
The Role of Sex Differences in Bone Health and Healing
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

CAM Model: Intriguing Natural Bioreactor for Sustainable Research and Reliable/Versatile Testing

Department of Biomedical, Metabolic and Neural Sciences, Section of Human Morphology, University of Modena and Reggio Emilia—Largo del Pozzo, 41124 Modena, Italy
*
Author to whom correspondence should be addressed.
Biology 2023, 12(9), 1219; https://doi.org/10.3390/biology12091219
Submission received: 3 August 2023 / Revised: 31 August 2023 / Accepted: 6 September 2023 / Published: 8 September 2023
(This article belongs to the Special Issue Multidisciplinary Insights on Bone Healing (Volume II))

Abstract

:

Simple Summary

The chicken embryo chorioallantoic membrane (CAM) is an in ovo model that has been known for years. It has mostly been used to test the characteristics of molecules and cell pellets and their potential interactions with vessels, particularly in cancer studies. Recently, we repurposed such a model by highlighting its ethical features, because, to a large extent, it can reduce the use of animal experimentation and produce rapid results. Its applications have multiplied in recent years, allowing for the development of more in-depth and comprehensive analyses and, thus, reducing the gap between in vitro and in vivo experimentation. Since the CAM model allows for the replacement, reduction, and refinement of preclinical experimentation (rules of the “3Rs”), it makes experimental research more sustainable and in line with animal welfare. The objective of this review is to illustrate the potential of the CAM assay, with a particular focus on the setup of organotypic cultures. This type of assay may be used as a preclinical model to assay recovery strategies for critically-sized bone injuries, i.e., severe fractures that do not spontaneously heal due to disruption of the vascular network and a large gap between the two bone stumps.

Abstract

We are witnessing the revival of the CAM model, which has already used been in the past by several researchers studying angiogenesis and anti-cancer drugs and now offers a refined model to fill, in the translational meaning, the gap between in vitro and in vivo studies. It can be used for a wide range of purposes, from testing cytotoxicity, pharmacokinetics, tumorigenesis, and invasion to the action mechanisms of molecules and validation of new materials from tissue engineering research. The CAM model is easy to use, with a fast outcome, and makes experimental research more sustainable since it allows us to replace, reduce, and refine pre-clinical experimentation (“3Rs” rules). This review aims to highlight some unique potential that the CAM-assay presents; in particular, the authors intend to use the CAM model in the future to verify, in a microenvironment comparable to in vivo conditions, albeit simplified, the angiogenic ability of functionalized 3D constructs to be used in regenerative medicine strategies in the recovery of skeletal injuries of critical size (CSD) that do not repair spontaneously. For this purpose, organotypic cultures will be planned on several CAMs set up in temporal sequences, and a sort of organ model for assessing CSD will be utilized in the CAM bioreactor rather than in vivo.

Graphical Abstract

1. Introduction

The field of clinical research has shown significant improvement in the therapeutic strategies, which are continuously developing to cope with the problems that an aging population imposes. The efficacy and the safety of these strategies need to be evaluated through extensive preclinical testing, including animal experimentation, which is essential for approval from regulatory office such as Food and Drug Administration (FDA) prior to being applied in clinical approaches. These necessities come in contrast with sustainable research; this is why there is a need for the development of alternative strategies to animal testing [1,2].
In recent years, thanks, in part, to some funding agencies that are more attentive to animal welfare, there has been much emphasis on projects that include the development of alternatives to animal experimentation, which, to date, appears to be a mandatory and necessary step for bringing new devices/therapies to market, but which, on the other hand, is too expensive and no longer sustainable. Alternatives proposed in the world of basic and clinical research include, for example, tissue cultures [3,4,5], 3D cultures [6,7,8,9], organoids [10,11], microfluidics such as organs-on-a-chip [12,13,14], and the use of chicken chorioallantoic membrane [15,16,17,18,19,20,21]. The latter is a technique that was developed in the past for angiogenesis assays because of its highly vascularized system, ease of use, inexpensiveness, and great ethical value. In the last decade, chorioallantoic membrane (CAM) assays have been re-discovered and used not only for traditional angiogenic assays [22,23] but also as non-innerved bioreactors and providers of rapidly growing vascular beds that mimic the blood supply for organ culture [24,25,26,27]. In addition, the embryo is not immunocompetent until 16–17th days of development; therefore, it cannot sustain rejection reactions [28,29,30]. The versatility of this model has made it widely used in recent years; in fact, this has opened new pathways to more sustainable, ethical, and animal-welfare-supportive research.
In particular, the field of bone regenerative medicine can benefit, for example, from the use of organ culture models with CAM. A fundamental prerequisite for the formation of new bone is the presence of a well-developed vascular bed that serves as a template for the generation of new bone thanks to the collaboration of the bone-forming cells [31,32,33,34,35,36]. The study of proangiogenic potential and its associated implications for tissue regeneration require complex in vivo models comprising all steps of the angiogenic process. The CAM model offers a simple, easily accessible, and inexpensive angiogenic screening tool compared to other animal models. In addition, the great ethical value of this in ovo model lies in the application of the 3Rs principle [37]: the possibility of being employed in multiple experiments, for different research fields, replaces the use of in vivo animal models for the experimental phases immediately following the in vitro experimentation (replace). A direct consequence of this is a reduction in the number of animals used during the experimental period (reduce). Finally, the use of a CAM assay allows researchers to minimize animal suffering, since the chick embryo does not exhibit nociception until day 11 of embryonic development (refine), as established by the National Institute of Health [38] as well as the Institutional Animal Care and Use Committee [39]. In addition, pain perception does not fully occur before the 15th day of embryonic development due to the immaturity of the portion of the central nervous system devoted to pain perception.
This review provides an overview of the uses of CAM assays in the two last decades and suggests the use of in ovo tests as an alternative to animal testing in preclinical studies; this could be a good solution to use in the field of regenerative medicine as a model for testing therapies for the resolution of critical bone fractures.

2. The CAM

The chicken embryo’s chorioallantoic membrane (CAM) is a highly vascularized extraembryonic structure that functions by exchanging gas and nutrients for the embryo during the entire period of its development; further, it is also responsible for calcium mobilization from eggshell to promote embryonic bone mineralization [40,41,42]. It originates from the fusion between the mesodermal layer of the allantois and the adjacent mesodermal layer of the chorion, forming a double layer structure with a rich vascular network (Figure 1C), connected with the embryonic circulation by two allantoic arteries and one allantoic vein [43]. The mature CAM morphologically resembles a “C” and is permeated by a fluid called allantoic fluid, which physiologically provides nutrients and carries waste substances out of the embryo [44,45]. Thanks to its features, CAM is a convenient and versatile biological instrument.
The timing of the embryo’s development was described by Hamburger and Hamilton in 1951, and was subdivided in 46 chronological stages (HH stages) by using specific characteristics that occur at each step of the chick’s development regardless of the exact age of the embryo [46]. However, chicks’ developmental time can be difficult to assess because the egg is internally fertilized and goes through a brief period of development; for this, actual incubation days are usually considered for experimental dating, assuming that embryonic development begins for all eggs simultaneously at the time of incubation at a constant temperature and humidity.
The CAM begins its formation at about day 3 of embryonic development (ED3) and reaches completion around ED9, which is precisely why its use requires that tests be set up no earlier than ED 8–9 [47]. No CAM experiments reach the hatching period of the eggs, which, for chickens, is set at day 21 of embryonic life; however, experimentation can be carried out either in ovo or ex ovo, Latin for “inside the egg” and “outside the egg”, respectively. Initially for both methods, fertilized eggs from avian species (i.e., quail, turkey, and duck [48,49,50,50,51,52,53,54]), most commonly chicken, are kept in a humidified incubator at a constant humidity of 45% and a temperature of 37 °C for up to 15 days, by which time experimentation is allowed without the need to seek ethics committee approval. Indeed, as stated earlier, as sanctioned by Institutional Animal Care and Use Committee (IACUC), the chicken embryo is not considered a living organism until the 17th day of embryonic life [38,39].
Within this two-week period, certain operations are performed to make the egg accessible: opening, insertion of materials/molecules to test, final observation, and tissue collection [55]. In the case of the ex ovo technique, at ED3, the eggshell is cracked and its contents are placed in a sterile container. Several authors have developed various methodologies: eggs in cubes [56], the use of petri plates [57], the use of plastic glasses with water [58]; weighing trays [59], etc. In the case of in ovo experimentation, at ED3, the egg can be opened by making a small window to access the shell after aspirating a few ml of albumen from the opposite pole to the air chamber, in order to preserve the CAM during the shell access procedure. In any methodology chosen, in the opening phase, the operator visualizes the viability of the embryo by verifying the palpitation of the heart and observing the embryonic morphology characterized by the presence of a cephalic bud, posterior part, neural tube, and the classic “spider” structure of the vessels disposed around the embryo and composed of the main veins and arteries feeding the embryo (Figure 1A). The CAM is planar and can be easily observed as a circled area highly vascularized around the embryo (Figure 1B).
The CAM is considered to lack immune competence until ED16–17. In fact, the lymphoid cells (i.e., mononuclear phagocytes, T and B cells) are present from ED11–12, but they are immature; therefore, the immune system is not active [60,61,62,63,64,65]. Since the CAM has a very dense capillary network, it is commonly used to study both new vessel formation (angiogenesis versus vasculogenesis) and its inhibition in response to different factors.
The main advantages of CAM include: low cost, function as a natural bioreactor and well-vascularized system; high reproducibility and reliability; the natural immune deficiency of non-sentient embryos, and no need for ethical approval (until ED17). On the other hand, as with all models, CAM displays some disadvantages: short time to allow for cell migration, similarity between pre-existing and newly-generated vessels, susceptibility to environmental changes and contaminations by fungi and bacteria, different metabolism with respect to humans, need for skill and practice to handle the model before hatching, and rapid morphological modifications during embryonic development. Other limitations of the CAM method include the variations due to the fertilization rate in different seasons, and some difficulties particularly related to ex ovo procedures and peculiar applications.
However, in recent years, the use of CAM has been greatly expanded, and there are now many applications.

3. The Application of CAM up to Now

The vascularized environment of the CAM of avian species offers the possibility to study a variety of molecules and materials; in particular, a CAM assay can be used to test pro/anti-angiogenic potential, to perform cancer studies, to test molecules and materials [66,67], to verify transplant reactions, and to test some drug effects.
The clinical research in the field of regenerative medicine typically involves an initial phase of in vitro testing, including tests of cytotoxicity, biocompatibility, and others, but some analyses need to be confirmed using animal models. In this situation, CAM can be used as an alternative to animal experimentation, since it is comparable to a natural “in ovo” bioreactor. This makes future research more sustainable and makes it possible to lower the costs of the pre-clinical phase and speed up the preliminary tests needed prior to animal testing (which is currently mandatory for product entry into the clinical phase).
The following paragraphs provide a brief overview of CAM’s uses for various experiments conducted in the last decade. Next, we will differentiate the past use of CAM with the perspective uses that can be made of it, such as the set-up, for example, of organotypic cultures [16].

3.1. Use of the Cam Assay for Cancer Studies

Jankovic B.D. et al. were among the first to assert that CAM, as a highly vascularized membrane, together with extracellular matrix (ECM) proteins, mimics the physiological environment of cancer [30]. Thus, the CAM assay is considered particularly suitable for studying the distinctive aspects of cancer, such as angiogenesis, invasion, metastasis formation, and cancer cell spread [68,69,70,71,72]. There are various advantages and disadvantages that make the use of CAM versus the use of animal models an alternative way to study tumors.
One advantage that the CAM model has over the animal model in the study of tumor invasion is the time required for the development of visible microtumors, which, in animal models, become evident only several weeks after cell transplantation, whereas in the CAM model, tumor growth can be observed as early as a few days after cell grafting [73].
The short timing of embryonic development is another benefit of using CAM, because it is possible to speed up and simplify data collection in the pre-clinical phase. Indeed, the entire period of embryonic development is faster compared to that of any animal model, and this allows for rapid morphological feedback, for example, on the development of the vascular network in response to different types of grafts. At the same time, the difficulty of distinguishing newly formed blood vessels from preexisting ones is the main disadvantage of using CAM in cancer research. Finally, among the disadvantages of using the CAM assay, there is the difficulty of maintaining a sterile system and avoiding environmental contamination [74].
As for its strengths, which are more numerous than its disadvantages, the CAM model is widely used for tumor grafting, which can be implanted on the membrane in various forms: patient-derived xenografts; solid biopsies; circulating cancer cells in suspension; or, most commonly, tumor cell lines. Patient-derived xenografts retain many of the biological features of primary tumors, and, therefore, by grafting them onto CAM, it becomes possible to investigate the genetic, protein, morphological, and pharmacological patterns, as well as cancer-specific immune evasion mechanisms [73,75,76,77]. By transplanting biopsies of mammalian tumors, it is possible to maintain the main features of primary tumors and to perform studies regarding cell polymorphisms, mechanisms of growth and angiogenesis, interaction with the extracellular matrix, and metastasis formation [78,79,80]. Grafting circulating tumor cells is useful for analyzing the aggressiveness and proliferation ability of primary tumors, with the aim of performing pre-clinical drug screening and discovering biomarkers [77].
Table 1 shows some of the several cell lines which have been implanted in CAM in the last decade.

3.2. Cancer Hallmarks Studied in CAM: Angiogenesis

The CAM model has been widely used in the past to study the hallmarks of cancer, such as angiogenesis, proliferation, and tumor invasion, as well as to analyze the conditions underlying cancer therapies [100]. The process of developing a vascular network that supplies nutrients and oxygen to tumor cells has obviously been the subject of multiple studies, because growing tumors take advantage of the host’s physiological angiogenesis and promote its exuberant development to secure adequate oxygen and nutrient supply, to dispose of waste products, and to facilitate the dissemination of tumor cells to other districts [101].
Tumor onset and progression take place in successive phases, one of which is the avascular phase, during which an “angiogenic switch” can be triggered, resulting in vascular branching and endothelial cell proliferation and allowing the tumor to grow while ensuring a sustained energy supply [102]. In this way, even an initially benign neoplasm can evolve and mutate, and vascular proliferation can allow for its development beyond its benign dimensions. The deregulation of angiogenesis is also an hallmark of cancer [103], and inhibition of the altered tumor angiogenic process has been utilized as a therapeutic strategy for a long time now [104].
After cell grafting on CAM, tumors become visible within 2–3 days and are readily supplied with CAM-derived blood vessels that penetrate deeply into the tissue. Several qualitative and quantitative approaches have been employed to assess the angiogenic response to different types of treatments [69].
Demcisakova Z. et al. validated angiogenetic potential by immunohistochemistry against embryonic endothelial markers such as WGA (wheat germ agglutinin) and SMA (smooth muscle actin), chicken-specific myofibroblast (α-SMA). Monoclonal antibodies specifically recognize chicken monocytes, macrophages, and interdigitating macrophage cells (KUL01); moreover, with RT-PCR, it is possible to quantify the gene expression of angiogenesis markers such as VEGF (vascular endothelial growth factor), FGF-2 (fibroblast growth factor-2), ANG-1 (angiopoietin-1), and HIF-1α (hypoxia-inducible factor 1-alpha) [105].
In other studies, the formation new vessels has been quantified through immunohistochemistry to chicken-specific CD34 (predominantly regarded as a marker of hematopoietic stem cells and hematopoietic progenitor cells) or using a particular lectin (biotinylated lens culinaris agglutinin) that binds specifically to endothelial cells of chicken veins, arteries, and capillaries. That hybridization was used to assess the angiogenesis that is generated at the intra-tumoral level after grafting osteosarcoma cells onto CAM [71,106]. The tumor supply system has been the object of many studies in which anti-angiogenic drugs and biomaterials have been used to slow down the tumor growth process. Some of these anti-angiogenic drugs tested in ovo have been molecules that inhibit the VEGF and the platelet-derived growth factor receptors [107,108,109]. Another anti-angiogenic mechanism tested in CAM involves microRNAs (miRNAs) that play a key role in gene expression [110,111]. Among these, microRNA-21 (miR-21) is an oncogenic miRNA [110], the overexpression of which can downregulate key tumor inhibitory proteins, such as programmed cell death protein [112], TNFα (tumor necrosis factor-α), ERK (extracellular signal-regulated kinase), and VEGF [113]. miRNA-based therapy can be considered as a knockdown of miR-21 expression, induction of tumor cell apoptosis, and suppression of tumor-associated angiogenesis [114,115].
Finally, in a study conducted by Tome Y., another strategy was tested in human osteosarcoma cells transplanted onto CAM, involving the echistatin. This cyclic peptide functions as an anti-angiogenic molecule by bonding to the integrin α v β 3, thus inhibiting it [116].

3.3. Cancer Hallmarks Studied in CAM: Metastatic Potential

For several years now, the CAM model has been recognized as a viable alternative to animal models for the characterization of tumors and for their metastatic potential [117]. The CAM model also allows for the potential development of metastases in all organs of the chicken embryo [73]. Along with this feature, the intrinsic aggressiveness of various tumor forms was found to have more explanatory elements in diagnostic and therapeutic phases. In this context, by supplying the chicken embryo’s circulatory system with blood and nutrients, CAM provides an ideal system by recreating the physiological microenvironment for the cell–cell and cell–matrix interaction studies that occur during the metastatic cascade [100]. After the injection of tumor cells into the circulatory system of the chicken embryo, metastatic potential can be assessed by tracking the mRNA levels of metastasizing cancer cells in chick embryos. Each metastatic site is analyzed from a morphological and invasive potential point of view [118]. Traditional morphological detection techniques used in animal models can also be used in the CAM model. Indeed, to identify cell morphology and location, tumor samples can be subjected to hematoxylin and eosin staining as shown by Shioda and coworkers, who detected colon cancer cells by labeling sections of embryonic organs with the anti-human pan cytokeratin antibody [118,119].
Cell invasiveness, moreover, can be monitored by labeling tumor cells with fluorescent molecules that allow for the detection of scattered cell colonies in the various embryonic body districts, and simultaneously labeling chicken blood vessels with a particular lectin (biotinylated lens culinaris agglutinin) [120,121]. Ranjan R.A. and his team compared two breast cancer cell lines, MCF-7 and MDA-MB 231, to study growth rates by morphological evaluation, proliferation by immunohistochemistry for the Ki-67 protein, aggressiveness by evaluating the mitotic rate and tumor budding, and, finally, cell spreading using the Alu-PCR assay [68].
The latter involves specific in situ hybridization of the repeated sequences in a human genome named Alu, which are present only in humans with a frequency of 5% [122,123]. Tissue sections are subjected to RT-PCR for Alu sequences and CR1 (Chicken Repeat-1) to make a quantitative assessment of the human tumor cells intravasating and disseminating into the chick embryo through the CAM. It also, at the same time, distinguishes human cells from chicken vessels [69,71,73,97,117,119,124,125,126]. The search for human gene sequences by RT-PCR for a certain determined gene has been a technique used for several years now; in fact, many years ago, Kobayashi and coworkers identified metastatic prostate cancer cells disseminated in the liver and femur of a chicken embryo by amplification of the human beta globin gene [127].

3.4. Tumor Therapy Test in CAM

The CAM model is a versatile, yet also relatively simple and low-cost, model that also allows for the screening of pharmacological or physical therapies within a short time. Moreover, the use of the CAM model can be considered a precision tool for medicine to be used in the search for tailored cancer drugs [128]. Drugs that inhibit tumor growth have been tested in CAM in two main ways: by injecting them into the chicken’s circulatory system or by using them as a treatment of tumor cells, seeded appropriately on CAM, as reported in the protocol developed by Kunz and his team [71].
Therefore, the CAM assay is a reference model for several therapeutic approaches, including various chemotherapeutics [86,97], targeted [129,130,131] and checkpoint therapies [132], oncolytic viruses [133], radiotherapy [134], molecules that block the cell cycle and induce apoptosis [135,136], and anti-angiogenesis drugs [137,138].

4. Use of the CAM Assay to Validate Scaffolds for Regenerative Purposes

Regenerative medicine, in recent years, is progressing toward new translational approaches based on the formulation and fabrication of Advanced Therapy Medical Products (ATMPs). It is, therefore, tissue engineering (TE), a branch of research that aims to produce constructs that are the results of a combination of cells, biomaterials, and biologically active molecules, in the form of scaffolds with the aim of repairing tissues by inducing their regeneration [139,140,141,142,143,144,145,146]. TE can be conducted ex vivo or in situ [147,148]: the first approach requires the seeding of donor stem cells onto a scaffold that is inserted into the affected tissue for the purpose of stimulating cell growth and differentiation [149,150,151,152]; the in situ method, on the other hand, avoids the step of seeding cells onto the scaffold and involves the fabrication of scaffolds that can adapt to tissue damage in terms of their size and shape. The latter contain biocompatible materials that can be implanted at the site of damaged tissue, where they attract the surrounding host cells necessary for healing to the repair site [145,147,148,153,154]. Specifically, among the components of TE constructs emerge biomaterials which hold many key characteristics for in vivo implantation into host tissues. These include biocompatibility to avoid the induction of an immune response, sterilizability to be safely incorporated into host tissues, biodegradability to be degraded by tissue cells into easily metabolized molecules from the tissue after performing their function, and bioactivity to stimulate tissue repair. Regardless of biochemical composition and biophysical properties, their most important feature is the interaction with the biological system in which they are embedded [155,156].
Biomaterials can be classified according to their origin. There are those of natural origin, such as chitosan, alginate, and cellulose [157,158], and those of synthetic origin, such as PLGA poly (lactic-co-glycolic acid), PCL (lactic-co-glycolic acid), PLA (polylactic acid), fibronectine, and polyurethane [158,159].
In this context, once again, the CAM model represents a natural bioreactor which can be used to test the main characteristics of biomaterials and the effects they have on the CAM, which represents a viable system. In recent years, the CAM assay become a popular approach in tissue engineering studies, in particular in the study of different tissue pathologies, such as those related to bone defects [64,160]. The chorioallantoic membrane allows for the observation of the effect that biomaterials have on the angiogenesis and tests their biocompatibility. Considering the central role that angiogenesis plays in tissue regeneration, the evaluation of the angiogenic potential of biomaterials has become a priority in TE, especially for bone TE [161,162,163,164,165].
The angiogenic potential and biocompatibility of several biomaterials have already been tested in CAM. Many of the biomaterials which have been tested in CAM are of synthetic origin; among them are hydrogels, which mimic extracellular matrix materials (ECM) due to their highly hydrated, permeable, and porous structures. They enable guided tissue regeneration by facilitating cellular activities, nutrient diffusion, and waste transfer [166]. The swelling and degradation ability of polymer matrix hydrogels makes them suitable for encapsulating and delivering numerous therapeutic agents, such as cells, growth factors, drugs, and genes, into tissue defects [167,168]. In addition, hydrogels are very often enriched with other molecules, such as, for example, acrylamine [169], heparin [170], and hyaluronic acid [171]. The latter is designed to treat periodontitis, a chronic biofilm-associated inflammatory disease of the tooth-supporting tissues that causes tooth loss. The scaffold developed by the team, based on controlled oxygen-releasing hyaluronic acid, is useful in avoiding a hypoxic environment that would compromise tissue regeneration [172,173]. Other biomaterials tested in CAM include bioplastics, which are eco-friendly materials used in bone tissue regeneration for their biocompatibility and biodegradability. Specifically, poly (3-hydroxybutyrate-co-3-hydroxyhexanoate) (PHBH) reinforced witch cellulose nanocrystals (CNCs) has been tested in CAM in the form of a porous scaffold. The CAM assay enabled the identification of the scaffold pore size, which is more optimal for endothelial cell colonization and blood vessel formation [67]. Bioactive glasses, in the TE field, have also received a significant amount of interest. These, enriched with biologically active ions of various kinds, such as boron, were the focus of research conducted by Decker and coworkers. They observed the influence of B-doping of bioactive glasses on the viability, osteogenic differentiation, and expression of osteogenic and angiogenic marker genes of bone marrow-derived mesenchymal stromal cells (BMSCs), in the presence of the B-BGs’ ionic dissolution products (IDPs); subsequently, they evaluated the influence of IDPs on chorioallanotic membrane angiogenesis [174]. In the same way as bioglass, synthetic hydroxyapatite (HA), which is a particular type of calcium phosphate, has been widely examined as a regeneration material because of its affinity to the main natural component of bone and its osteoconductivity and bioactivity [175,176,177,178,179]. In this regard, HA formulated with other biomaterials, such as biopolymers, demonstrates remarkable vasculogenesis, as is evident from CAM testing conducted in recent studies focused on finding viable regenerative strategies for the orbital floor [159]. Other biopolymers also fit into this context: Demsisakova et al. developed a scaffold consisting of the biopolymer polyhydroxybutyrate (PHB) combined with chitosan (CHIT). Also, using the CAM assay, they demonstrated that (PHB)/(CHIT) has strong endogenous angiogenic potential and could be a promising biomaterial for the treatment of hard tissue defects [105]. The most significant advantage of using CAM in studying the properties of biomaterials is that the CAM allows for the development and branching of the vascular network on the implanted scaffolds, mimicking what should occur in tissue in vivo. In this regard, the porosity and pore size of the scaffold play key roles in vascular infiltration and osteogenic differentiation [180,181,182]; therefore, the challenge for researchers seems to be to formulate ever-new constructs that have better porosity and efficiency in TE.

5. Use of CAM to Set-Up Organotypic Culture

All the advantages of CAM also make it an attractive model for tissue engineering approaches. The membrane, during the developmental stages of the chick embryo, provides a naturally immunocompromised host and a rapidly growing vascular bed that lacks a nervous system and, therefore, provides a less sentient alternative for animal research. This allows for in vivo implantation of organs and represents a model for xenografts developing organotypic cultures [73,183]. This provides a solution to the most important limitation of the ex vivo organotypic culture: the lack of blood, immunity cells, and bone cells [184,185].
Given that, especially in the field of bone tissue engineering, it is possible to use both the embryo and the membrane itself, Blake et al., on the 18th day of embryo development, harvested the femur of the embryo, which they then implanted onto the CAM after causing a fracture [184].
As shown by Aldamash A. et al. [186] and Marshall et al. [187], the use of the chicken embryo femur also had another application; the works of these two researcher groups aimed to examine the differentiation potential of specific cells, such as human bone marrow stromal cells (HBMSCs) and human neonatal foreskin stromal cells (hNSSCs), alone or in combination with human umbilical vein endothelial cells (HUVECs), under experimental conditions for tissue regeneration. The authors took the chicken embryo femur, caused a fracture, and cultured it with different cell types to test their differentiation potential as well as, in the case of hNSSCs and HUVECs, their angiogenic potential.
Although the CAM model per se allows for short experimental times (from day x to day y), it is also possible to overcome this time limit by performing transplantation from one chorion allantoic membrane to another ex vivo of organotypic cultures or biomaterials to be tested on CAM; of course, in case of the need for increased experimental time, one must be careful not to damage the grafted samples/scaffolds in transport from one CAM to the other. In a recent study, Feder et al. set up a protocol whereby it is possible to graft onto the CAM various sections of osteosarcoma tissue taken from rats and mice; then, they transplanted them onto another membrane five to seven consecutive times, enabling further experimental analyses [188].
Another insight about CAM’s applications in this field is provided by Kanczler and his team, who devised a critically-sized chick femur defect model, which has been used to evaluate different types of molecules engaged in bone tissue engineering. Specifically, CAM is used to test the effects of different factors and proteins involved in healing bone defects, such as bone morphogenetic protein-2 (BMP-2), vitamin D3, parathyroid hormone (PTH), and parathyroid-hormone-related protein (PTHRP), to assess the potential of osseointegration of scaffolds and to evaluate their performance before using them in in vivo studies [189,190,191].
Other studies report how CAM can also be used for transplanting sections of organs or organoids from murine embryos, such as, for example, the kidney. Embryonic kidneys were taken at ED11.5, transplanted onto CAM at ED8, and then cultivated for 7 days; subsequently, the murine embryonic kidney, grown on CAM, underwent immunohistochemistry for endothelial growth markers, highlighting the anastomosis between the blood vessels of CAM and those of the murine kidney [192].

6. Discussion and Conclusions

As described above, there is ample evidence that the use of CAM in numerous research areas is effective for research and clinical studies. A positive fact is that the CAM model, inspired by the 3R concept, is a viable alternative to classical animal experimentation, which is no longer sustainable without ethical limitations/controls and which, in the future, will have to be replaced with (or must be accompanied by) alternatives that are more advantageous in economic, ethical, and experimental terms. It has already been pointed out that research in the field of oncology and into biomaterials for regenerative medicine are highly developed areas, in which more resources have been invested in the development of techniques and strategies for the use of CAM corresponding to the demands of research. In addition, other applications of CAM are cautiously being developed in order to use this powerful vascularized natural bioreactor for the accomplishment of organ cultures, which could replace the early stages of experimentation and are currently performed exclusively on animal models, thus decreasing the negative impact of research on animal welfare.
This review has highlighted the most significant scientific studies from the last decade to underline the current importance of this alternative model to animal experimentation. Finally, we would like to point out that the interest in the use of CAM has recently materialized, with the 1st International Conference on CAM held in February 2022, which brought together highly prestigious scholars from all over the world.

7. Future Directions

In the future, it is to be hoped that even more standardized techniques will be developed for setting up CAM testing services for large research macro-areas in order to meet all scientific demands. In addition, more publicity should be given to this alternative model to in vivo testing due to all of the advantages and properties described above. The possibility of setting up organ cultures on CAM is attractive, useful, and innovative. At present, the use of this tool is not widespread, and the development of methodologies for the use of CAM for organ cultures is slow. In the coming years, it is hoped that more and more research groups will devote themselves to the implementation of this organ culture model in order to test various molecules and clinical strategies from a translational perspective.

Author Contributions

Writing—original draft preparation, M.C. and F.S.; review, editing, and supervision, C.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

Thanks are due to Alberto Martelli of the University of Bologna for providing the BioRender software.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hippenstiel, S.; Thöne-Reineke, C.; Kurreck, J. Animal Experiments: EU Is Pushing to Find Substitutes Fast. Nature 2021, 600, 37. [Google Scholar] [CrossRef] [PubMed]
  2. Fentem, J.; Malcomber, I.; Maxwell, G.; Westmoreland, C. Upholding the EU’s Commitment to ‘Animal Testing as a Last Resort’ Under REACH Requires a Paradigm Shift in How We Assess Chemical Safety to Close the Gap Between Regulatory Testing and Modern Safety Science. ATLA Altern. Lab. Anim. 2021, 49, 122–132. [Google Scholar] [CrossRef] [PubMed]
  3. Aguilar, A.H.; Smith, L.; Owens, D.; Quelch, R.; Przyborski, S. Recreating Tissue Structures Representative of Teratomas In Vitro Using a Combination of 3D Cell Culture Technology and Human Embryonic Stem Cells. Bioengineering 2022, 9, 185. [Google Scholar] [CrossRef] [PubMed]
  4. Bédard, P.; Gauvin, S.; Ferland, K.; Caneparo, C.; Pellerin, È.; Chabaud, S.; Bolduc, S. Bioengineering Innovative Human Three-Dimensional Tissue-Engineered Models as an Alternative to Animal Testing. Bioengineering 2020, 7, 115. [Google Scholar] [CrossRef]
  5. Darling, N.J.; Mobbs, C.L.; González-Hau, A.L.; Freer, M.; Przyborski, S. Bioengineering Novel in Vitro Co-Culture Models That Represent the Human Intestinal Mucosa With Improved Caco-2 Structure and Barrier Function. Front. Bioeng. Biotechnol. 2020, 8, 992. [Google Scholar] [CrossRef]
  6. Knight, E.; Murray, B.; Carnachan, R.; Przyborski, S. Alvetex®: Polystyrene Scaffold Technology for Routine Three Dimensional Cell Culture. In Methods in Molecular Biology; Humana Press Inc.: Totowa, NJ, USA, 2011; Volume 695, pp. 323–340. [Google Scholar]
  7. Golinelli, G.; Talami, R.; Frabetti, S.; Candini, O.; Grisendi, G.; Spano, C.; Chiavelli, C.; Arnaud, G.F.; Mari, G.; Dominici, M. A 3D Platform to Investigate Dynamic Cell-to-Cell Interactions Between Tumor Cells and Mesenchymal Progenitors. Front. Cell Dev. Biol. 2022, 9, 767253. [Google Scholar] [CrossRef]
  8. Flagelli, A.; Candini, O.; Frabetti, S.; Dominici, M.; Giardino, L.; Calzà, L.; Baldassarro, V.A. A Novel Three-Dimensional Culture Device Favors a Myelinating Morphology of Neural Stem Cell-Derived Oligodendrocytes. Front. Cell Dev. Biol. 2021, 9, 759982. [Google Scholar] [CrossRef]
  9. Shahin-Shamsabadi, A.; Selvaganapathy, P.R. Tissue-in-a-Tube: Three-Dimensional In Vitro Tissue Constructs with Integrated Multimodal Environmental Stimulation. Mater. Today Bio 2020, 7, 100070. [Google Scholar] [CrossRef]
  10. Mukhopadhyay, C.; Paul, M.K. Organoid-Based 3D In Vitro Microphysiological Systems as Alternatives to Animal Experimentation for Preclinical and Clinical Research. Arch. Toxicol. 2023, 97, 1429–1431. [Google Scholar] [CrossRef]
  11. Puschhof, J.; Pleguezuelos-Manzano, C.; Clevers, H. Organoids and Organs-on-Chips: Insights into Human Gut-Microbe Interactions. Cell Host Microbe 2021, 29, 867–878. [Google Scholar] [CrossRef]
  12. Low, L.A.; Mummery, C.; Berridge, B.R.; Austin, C.P.; Tagle, D.A. Organs-on-Chips: Into the next Decade. Nat. Rev. Drug Discov. 2021, 20, 345–361. [Google Scholar] [CrossRef] [PubMed]
  13. Tao, T.; Wang, Y.; Chen, W.; Li, Z.; Su, W.; Guo, Y.; Deng, P.; Qin, J. Engineering Human Islet Organoids from IPSCs Using an Organ-on-Chip Platform. Lab Chip 2019, 19, 948–958. [Google Scholar] [CrossRef] [PubMed]
  14. Manafi, N.; Shokri, F.; Achberger, K.; Hirayama, M.; Mohammadi, M.H.; Noorizadeh, F.; Hong, J.; Liebau, S.; Tsuji, T.; Quinn, P.M.J.; et al. Organoids and Organ Chips in Ophthalmology. Ocul. Surf. 2021, 19, 1–15. [Google Scholar] [CrossRef] [PubMed]
  15. Chen, L.; Wang, S.; Feng, Y.; Zhang, J.; Du, Y.; Zhang, J.; Van Ongeval, C.; Ni, Y.; Li, Y. Cells Utilisation of Chick Embryo Chorioallantoic Membrane as a Model Platform for Imaging-Navigated Biomedical Research. Cells 2021, 10, 463. [Google Scholar] [CrossRef] [PubMed]
  16. Ribatti, D. Two New Applications in the Study of Angiogenesis the CAM Assay: Acellular Scaffolds and Organoids. Microvasc. Res. 2022, 140, 104304. [Google Scholar] [CrossRef] [PubMed]
  17. Fonseca, B.B.; da Silva, M.V.; de Morais Ribeiro, L.N. The Chicken Embryo as an In Vivo Experimental Model for Drug Testing: Advantages and Limitations. Lab. Anim. 2021, 50, 138–139. [Google Scholar] [CrossRef]
  18. Dadhich, P.; Das, B.; Pal, P.; Srivas, P.K.; Dutta, J.; Ray, S.; Dhara, S. A Simple Approach for an Eggshell-Based 3D-Printed Osteoinductive Multiphasic Calcium Phosphate Scaffold. ACS Appl. Mater. Interfaces 2016, 8, 11910–11924. [Google Scholar] [CrossRef]
  19. Burgio, F.; Rimmer, N.; Pieles, U.; Buschmann, J.; Beaufils-Hugot, M. Characterization and in Ovo Vascularization of a 3D-Printed Hydroxyapatite Scaffold with Different Extracellular Matrix Coatings under Perfusion Culture. Biol. Open 2018, 7, bio034488. [Google Scholar] [CrossRef]
  20. Baiguera, S.; Macchiarini, P.; Ribatti, D. Chorioallantoic Membrane for In Vivo Investigation of Tissue-Engineered Construct Biocompatibility. J. Biomed. Mater. Res. B Appl. Biomater. 2012, 100, 1425–1434. [Google Scholar] [CrossRef]
  21. Yalcin, H.C.; Shekhar, A.; Rane, A.A.; Butcher, J.T. An Ex-Ovo Chicken Embryo Culture System Suitable for Imaging and Microsurgery Applications. J. Vis. Exp. 2010, 44, e2154. [Google Scholar] [CrossRef]
  22. Moreno-Jiménez, I.; Kanczler, J.M.; Hulsart-Billstrom, G.; Inglis, S.; Oreffo, R.O.C. The Chorioallantoic Membrane Assay for Biomaterial Testing in Tissue Engineering: A Short-Term In Vivo Preclinical Model. Tissue Eng. Part. C Methods 2017, 23, 938–952. [Google Scholar] [CrossRef] [PubMed]
  23. Ribatti, D.; Nico, B.; Vacca, A.; Roncali, L.; Burri, P.H.; Djonov, V. Chorioallantoic Membrane Capillary Bed: A Useful Target for Studying Angiogenesis and Anti-Angiogenesis In Vivo. Anat. Rec. 2001, 264, 317–324. [Google Scholar] [CrossRef] [PubMed]
  24. Isachenko, V.; Mallmann, P.; Petrunkina, A.M.; Rahimi, G.; Nawroth, F.; Hancke, K.; Felberbaum, R.; Genze, F.; Damjanoski, I.; Isachenko, E. Comparison of In Vitro- and Chorioallantoic Membrane (CAM)-Culture Systems for Cryopreserved Medulla-Contained Human Ovarian Tissue. PLoS ONE 2012, 7, e32549. [Google Scholar] [CrossRef]
  25. Moreno-Jiménez, I.; Lanham, S.A.; Kanczler, J.M.; Hulsart-Billstrom, G.; Evans, N.D.; Oreffo, R.O.C. Remodelling of Human Bone on the Chorioallantoic Membrane of the Chicken Egg: De Novo Bone Formation and Resorption. J. Tissue Eng. Regen. Med. 2018, 12, 1877–1890. [Google Scholar] [CrossRef]
  26. Fazely, F.; Moses, D.C.; Ledinko, N. Effects of Retinoids on Invasion of Organ Cultures of Chick Chorioallantoic Membrane by Adenovirus Transformed Cells. In Vitro Cell. Dev. Biol. 1985, 21, 409–414. [Google Scholar] [CrossRef] [PubMed]
  27. Martinez-Madrid, B.; Donnez, J.; Van Eyck, A.S.; Veiga-Lopez, A.; Dolmans, M.M.; Van Langendonckt, A. Chick Embryo Chorioallantoic Membrane (CAM) Model: A Useful Tool to Study Short-Term Transplantation of Cryopreserved Human Ovarian Tissue. Fertil. Steril. 2009, 91, 285–292. [Google Scholar] [CrossRef]
  28. Leene, W.; Duyzings, M.J.M.; Van Steeg, C. Lymphoid Stem Cell Identification in the Developing Thymus and Bursa of Fabricius of the Chick. Z. Zellforsch. 1973, 136, 521–533. [Google Scholar] [CrossRef]
  29. Ribatti, D. The Chick Embryo Chorioallantoic Membrane in the Study of Angiogenesis and Metastasis; Springer: Berlin/Heidelberg, Germany, 2010; ISBN 978-90-481-3843-2. [Google Scholar]
  30. Jankovic, B.D.; Isakovic, K.; Lukic, M.L.; Vujanovic, N.L.; Petrovic, S.; Markovic, B.M. Immunological Capacity of the Chicken Embryo. I. Relationship between the Maturation of Lymphoid Tissues and the Occurrence of Cell-Mediated Immunity in the Developing Chicken Embryo. Immunology 1975, 29, 497–508. [Google Scholar]
  31. Genova, T.; Petrillo, S.; Zicola, E.; Roato, I.; Ferracini, R.; Tolosano, E.; Altruda, F.; Carossa, S.; Mussano, F.; Munaron, L. The Crosstalk between Osteodifferentiating Stem Cells and Endothelial Cells Promotes Angiogenesis and Bone Formation. Front. Physiol. 2019, 10, 1291. [Google Scholar] [CrossRef]
  32. Kanczler, J.M.; Oreffo, R.O.C. Osteogenesis and Angiogenesis: The Potential for Engineering Bone. Eur. Cell Mater. 2008, 15, 100–114. [Google Scholar] [CrossRef]
  33. Portal-Núñez, S.; Lozano, D.; Esbrit, P. Role of Angiogenesis on Bone Formation. Histol. Histopathol. 2012, 27, 559–566. [Google Scholar] [CrossRef] [PubMed]
  34. Checchi, M.; Stanzani, V.; Truocchio, S.; Corradini, M.; Ferretti, M.; Palumbo, C. From Morphological Basic Research to Proposals for Regenerative Medicine through a Translational Perspective. Ital. J. Anat. Embryol. 2022, 126, 139–145. [Google Scholar] [CrossRef]
  35. Palumbo, C.; Cavani, F.; Sena, P.; Benincasa, M.; Ferretti, M. Osteocyte Apoptosis and Absence of Bone Remodeling in Human Auditory Ossicles and Scleral Ossicles of Lower Vertebrates: A Mere Coincidence or Linked Processes? Calcif. Tissue Int. 2012, 90, 211–218. [Google Scholar] [CrossRef] [PubMed]
  36. Ferretti, M.; Palumbo, C. Static Osteogenesis versus Dynamic Osteogenesis: A Comparison between Two Different Types of Bone Formation. Appl. Sci. 2021, 11, 2025. [Google Scholar] [CrossRef]
  37. Russell, W.M.S.; Burch, R.L. The Principles of Humane Experimental Technique; Methuen: London, UK, 1959. [Google Scholar]
  38. National Institutes of Health. The Public Health Service Responds to Commonly Asked Questions. ILAR J. 1991, 33, 68–70. [Google Scholar] [CrossRef]
  39. Institutional Animal Care and Use Committee (IACUC). Policy for Use of Avian Embryos; Brown University: Schaumburg, IL, USA, 2019. [Google Scholar]
  40. Elaroussi, M.A.; DeLuca, H.F. Calcium Uptake by Chorioallantoic Membrane: Effects of Vitamins D and K. Endocrinol. Metab. 1994, 267, E837–E841. [Google Scholar] [CrossRef]
  41. Tuan, R.; Ono, T. Regulation of Extraembryonic Calcium Mobilization by the Developing Chick Embryo. J. Embryol. Exp. Morphol. 1986, 97, 63–74. [Google Scholar] [CrossRef]
  42. Packard, M.J. Mobilization of Shell Calcium by Chick Chorioallantoic Membrane In Vitro. J. Exp. Biol. 1994, 190, 141–153. [Google Scholar] [CrossRef]
  43. Maibier, M.; Reglin, B.; Nitzsche, B.; Xiang, W.; Rong, W.W.; Hoffmann, B.; Djonov, V.; Secomb, T.W.; Pries, A.R. Structure and Hemodynamics of Vascular Networks in the Chorioallantoic Membrane of the Chicken. Am. J. Physiol. Heart Circ. Physiol. 2016, 311, H913–H926. [Google Scholar] [CrossRef]
  44. Li, Y.; Qu, H.; Ji, J.; Wang, Y.; Liu, T.; He, J.; Wang, J.; Shu, D.; Luo, C. Characterization of the Exosomes in the Allantoic Fluid of the Chicken Embryo. Can. J. Anim. Sci. 2021, 101, 307–317. [Google Scholar] [CrossRef]
  45. Da Silva, M.; Labas, V.; Nys, Y.; Rehault-Godbert, S. Investigating Proteins and Proteases Composing Amniotic and Allantoic Fluids during Chicken Embryonic Development. Poult. Sci. 2017, 96, 2931–2941. [Google Scholar] [CrossRef]
  46. Hamburger, V.; Hamilton, H.L. A Series of Normal Stages in the Development of the Chick Embryo. J. Morphol. 1951, 88, 49–92. [Google Scholar] [CrossRef]
  47. Crespo, P.; Casar, B. The Chick Embryo Chorioallantoic Membrane as an In Vivo Model to Study Metastasis. Bio. Protoc. 2016, 6, e1962. [Google Scholar] [CrossRef]
  48. Lazarovici, P.; Lahiani, A.; Gincberg, G.; Haham, D.; Marcinkiewicz, C.; Lelkes, P.I. Nerve Growth Factor-Induced Angiogenesis: 2. The Quail Chorioallantoic Membrane Assay. In Neurotrophic Factors: Methods and Protocols; Skaper, S.D., Ed.; Springer: New York, NY, USA, 2018; Volume 1727, pp. 251–259. ISBN 978-1-4939-7571-6. [Google Scholar]
  49. Parsons-Wingerter, P.; Lwai, B.; Che Yang, M.; Elliott, K.E.; Milaninia, A.; Redlitz, A.; Clark, J.I.; Helene Sage, E. A Novel Assay of Angiogenesis in the Quail Chorioallantoic Membrane: Stimulation by BFGF and Inhibition by Angiostatin According to Fractal Dimension and Grid Intersection. Microvasc. Res. 1998, 55, 201–214. [Google Scholar] [CrossRef]
  50. Kundeková, B.; Máčajová, M.; Meta, M.; Čavarga, I.; Bilčík, B. Chorioallantoic Membrane Models of Various Avian Species: Differences and Applications. Biology 2021, 10, 301. [Google Scholar] [CrossRef]
  51. Rasmussen, S.V.; Berlow, N.E.; Price, L.H.; Mansoor, A.; Cairo, S.; Rugonyi, S.; Keller, C. Preclinical Therapeutics Ex Ovo Quail Eggs as a Biomimetic Automation-Ready Xenograft Platform. Sci. Rep. 2021, 11, 23302. [Google Scholar] [CrossRef]
  52. Lusimbo, W.S.; Leighton, F.A.; Wobeser, G.A. Histology and Ultrastructure of the Chorioallantoic Membrane of the Mallard Duck (Anas Platyrhynchos). Anat. Rec. 2000, 259, 25–34. [Google Scholar] [CrossRef]
  53. Buhr, C.R.; Wiesmann, N.; Tanner, R.C.; Brieger, J.; Eckrich, J. The Chorioallantoic Membrane Assay in Nanotoxicological Research—An Alternative for In Vivo Experimentation. Nanomaterials 2020, 10, 2328. [Google Scholar] [CrossRef]
  54. Longenecker, B.M.; Pazderka, F.; Stone, H.S.; Gavora, J.S.; Ruth, R.F. In Ovo Assay for Marek’s Disease Virus and Turkey Herpesvirus. Infect. Immun. 1975, 11, 922–931. [Google Scholar] [CrossRef]
  55. Janser, F.; Ney, P.; Pinto, M.; Langer, R.; Tschan, M. The Chick Chorioallantoic Membrane (CAM) Assay as a Three-Dimensional Model to Study Autophagy in Cancer Cells. Bio. Protoc. 2019, 9, e3290. [Google Scholar] [CrossRef]
  56. Huang, W.; Arai, F.; Kawahara, T. Egg-in-Cube: Design and Fabrication of a Novel Artificial Eggshell with Functionalized Surface. PLoS ONE 2015, 10, e0118624. [Google Scholar] [CrossRef]
  57. Dohle, D.S.; Pasa, S.D.; Gustmann, S.; Laub, M.; Wissler, J.H.; Jennissen, H.P.; Dünker, N. Chick Ex Ovo Culture and Ex Ovo CAM Assay: How It Really Works. J. Vis. Exp. 2010, 33, e1620. [Google Scholar] [CrossRef]
  58. García-Gareta, E.; Binkowska, J.; Kohli, N.; Sharma, V. Towards the Development of a Novel Ex Ovo Model of Infection to Pre-Screen Biomaterials Intended for Treating Chronic Wounds. J. Funct. Biomater. 2020, 11, 37. [Google Scholar] [CrossRef]
  59. Winter, R.; Dungel, P.; Reischies, F.M.J.; Rohringer, S.; Slezak, P.; Smolle, C.; Spendel, S.; Kamolz, L.P.; Ghaffari-Tabrizi-Wizsy, N.; Schicho, K. Photobiomodulation (PBM) Promotes Angiogenesis in-Vitro and in Chick Embryo Chorioallantoic Membrane Model. Sci. Rep. 2018, 8, 17080. [Google Scholar] [CrossRef]
  60. Gómez Del Moral, M.; Fonfría, J.; Varas, A.; Jiménez, E.; Moreno, J.; Zapata, A.G. Appearance and Development of Lymphoid Cells in the Chicken (Gallus Gallus) Caecal Tonsil. Anat. Rec. 1998, 250, 182–189. [Google Scholar] [CrossRef]
  61. Kunzi-Rapp, K.; Rück, A.; Kaufmann, R. Characterization of the Chick Chorioallantoic Membrane Model as a Short-Term In Vivo System for Human Skin. Arch. Dermatol. Res. 1999, 291, 290–295. [Google Scholar] [CrossRef] [PubMed]
  62. Ribatti, D.; Nico, B.; Vacca, A.; Presta, M. The Gelatin Sponge-Chorioallantoic Membrane Assay. Nat. Protoc. 2006, 1, 85–91. [Google Scholar] [CrossRef] [PubMed]
  63. Nowak-Sliwinska, P.; Segura, T.; Iruela-Arispe, M.L. The Chicken Chorioallantoic Membrane Model in Biology, Medicine and Bioengineering. Angiogenesis 2014, 17, 779–804. [Google Scholar] [CrossRef]
  64. Moreno-Jiménez, I.; Hulsart-Billstrom, G.; Lanham, S.A.; Janeczek, A.A.; Kontouli, N.; Kanczler, J.M.; Evans, N.D.; Oreffo, R.O.C. The Chorioallantoic Membrane (CAM) Assay for the Study of Human Bone Regeneration: A Refinement Animal Model for Tissue Engineering. Sci. Rep. 2016, 6, 32168. [Google Scholar] [CrossRef]
  65. DeBord, L.C.; Pathak, R.R.; Villaneuva, M.; Liu, H.-C.; Harrington, D.A.; Yu, W.; Lewis, M.T.; Sikora, A.G. The Chick Chorioallantoic Membrane (CAM) as a Versatile Patient-Derived Xenograft (PDX) Platform for Precision Medicine and Preclinical Research. Am. J. Cancer Res. 2018, 8, 1642–1660. [Google Scholar]
  66. Checchi, M.; Bertacchini, J.; Cavani, F.; Magarò, M.S.; Reggiani Bonetti, L.; Pugliese, G.R.; Tamma, R.; Ribatti, D.; Maurel, D.B.; Palumbo, C. Scleral Ossicles: Angiogenic Scaffolds, a Novel Biomaterial for Regenerative Medicine Applications. Biomater. Sci. 2020, 8, 413–425. [Google Scholar] [CrossRef] [PubMed]
  67. Stanzani, V.; Giubilini, A.; Checchi, M.; Bondioli, F.; Messori, M.; Palumbo, C. Eco-Sustainable Approaches in Bone Tissue Engineering: Evaluating the Angiogenic Potential of Different Poly(3-Hydroxybutyrate-Co-3-Hydroxyhexanoate)–Nanocellulose Composites with the Chorioallantoic Membrane Assay. Adv. Eng. Mater. 2023, 25, 2200934. [Google Scholar] [CrossRef]
  68. Ranjan, R.A.; Muenzner, J.K.; Kunze, P.; Geppert, C.I.; Ruebner, M.; Huebner, H.; Fasching, P.A.; Beckmann, M.W.; Bäuerle, T.; Hartmann, A.; et al. The Chorioallantoic Membrane Xenograft Assay as a Reliable Model for Investigating the Biology of Breast Cancer. Cancers 2023, 15, 1704. [Google Scholar] [CrossRef] [PubMed]
  69. Miebach, L.; Berner, J.; Bekeschus, S. In Ovo Model in Cancer Research and Tumor Immunology. Front. Immunol. 2022, 13, 1006064. [Google Scholar] [CrossRef]
  70. Schneider-Stock, R.; Ribatti, D. The CAM Assay as an Alternative In Vivo Model for Drug Testing. In Handbook of Experimental Pharmacology; Springer Science and Business Media Deutschland GmbH: Berlin/Heidelberg, Germany, 2020; Volume 265, pp. 303–323. [Google Scholar]
  71. Kunz, P.; Schenker, A.; Sähr, H.; Lehner, B.; Fellenberg, J. Optimization of the Chicken Chorioallantoic Membrane Assay as Reliable In Vivo Model for the Analysis of Osteosarcoma. PLoS ONE 2019, 14, e0215312. [Google Scholar] [CrossRef]
  72. Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef]
  73. Chu, P.Y.; Koh, A.P.F.; Antony, J.; Huang, R.Y.J. Applications of the Chick Chorioallantoic Membrane as an Alternative Model for Cancer Studies. Cells Tissues Organs 2022, 211, 222–237. [Google Scholar] [CrossRef]
  74. Doege, A.; Steens, R.; Dünker, N.; Busch, M.A. Retinoblastoma Cell Growth In Vitro and Tumor Formation In Ovo—Influence of Different Culture Conditions. Methods Protoc. 2022, 5, 21. [Google Scholar] [CrossRef]
  75. Thelen, M.; Wennhold, K.; Lehmann, J.; Garcia-Marquez, M.; Klein, S.; Kochen, E.; Lohneis, P.; Lechner, A.; Wagener-Ryczek, S.; Plum, P.S.; et al. Cancer-Specific Immune Evasion and Substantial Heterogeneity within Cancer Types Provide Evidence for Personalized Immunotherapy. NPJ Precis. Oncol. 2021, 5, 52. [Google Scholar] [CrossRef]
  76. Fischer, D.; Fluegen, G.; Garcia, P.; Ghaffari-Tabrizi-Wizsy, N.; Gribaldo, L.; Huang, R.Y.J.; Rasche, V.; Ribatti, D.; Rousset, X.; Pinto, M.T.; et al. The CAM Model—Q&A with Experts. Cancers 2023, 15, 191. [Google Scholar]
  77. Pizon, M.; Schott, D.; Pachmann, U.; Schobert, R.; Pizon, M.; Wozniak, M.; Bobinski, R.; Pachmann, K. Chick Chorioallantoic Membrane (CAM) Assays as a Model of Patient-Derived Xenografts from Circulating Cancer Stem Cells (CCSCs) in Breast Cancer Patients. Cancers 2022, 14, 1476. [Google Scholar] [CrossRef] [PubMed]
  78. Hu, J.; Ishihara, M.; Chin, A.I.; Wu, L. Establishment of Xenografts of Urological Cancers on Chicken Chorioallantoic Membrane (CAM) to Study Metastasis. Precis. Clin. Med. 2019, 2, 140–151. [Google Scholar] [CrossRef] [PubMed]
  79. Xiao, X.; Zhou, X.; Ming, H.; Zhang, J.; Huang, G.; Zhang, Z.; Li, P. Chick Chorioallantoic Membrane Assay: A 3D Animal Model for Study of Human Nasopharyngeal Carcinoma. PLoS ONE 2015, 10, e0130935. [Google Scholar] [CrossRef] [PubMed]
  80. Balčiūnienė, N.; Tamašauskas, A.; Valančiūtė, A.; Deltuva, V.; Vaitiekaitis, G.; Gudinavičienė, I.; Weis, J.; Graf Von Keyserlingk, D.; Balčiūnienė, N. Histology of Human Glioblastoma Transplanted on Chicken Chorioallantoic Membrane. Medicina 2009, 45, 123. [Google Scholar] [CrossRef] [PubMed]
  81. Vézina-Dawod, S.; Perreault, M.; Guay, L.D.; Gerber, N.; Gobeil, S.; Biron, E. Synthesis and Biological Evaluation of Novel 1,4-Benzodiazepin-3-One Derivatives as Potential Antitumor Agents against Prostate Cancer. Bioorg. Med. Chem. 2021, 45, 116314. [Google Scholar] [CrossRef]
  82. Goehringer, N.; Biersack, B.; Peng, Y.; Schobert, R.; Herling, M.; Ma, A.; Nitzsche, B.; Höpfner, M. Anticancer Activity and Mechanisms of Action of New Chimeric EGFR/HDAC-Inhibitors. Int. J. Mol. Sci. 2021, 22, 8432. [Google Scholar] [CrossRef]
  83. Miebach, L.; Freund, E.; Horn, S.; Niessner, F.; Sagwal, S.K.; von Woedtke, T.; Emmert, S.; Weltmann, K.D.; Clemen, R.; Schmidt, A.; et al. Tumor Cytotoxicity and Immunogenicity of a Novel V-Jet Neon Plasma Source Compared to the KINPen. Sci. Rep. 2021, 11, 136. [Google Scholar] [CrossRef]
  84. Liedtke, K.R.; Freund, E.; Hermes, M.; Oswald, S.; Heidecke, C.D.; Partecke, L.I.; Bekeschus, S. Gas Plasma-Conditioned Ringer’s Lactate Enhances the Cytotoxic Activity of Cisplatin and Gemcitabine in Pancreatic Cancer In Vitro and In Ovo. Cancers 2020, 12, 123. [Google Scholar] [CrossRef]
  85. Privat-Maldonado, A.; Verloy, R.; Cardenas Delahoz, E.; Lin, A.; Vanlanduit, S.; Smits, E.; Bogaerts, A. Cold Atmospheric Plasma Does Not Affect Stellate Cells Phenotype in Pancreatic Cancer Tissue in Ovo. Int. J. Mol. Sci. 2022, 23, 1954. [Google Scholar] [CrossRef]
  86. Busch, M.; Papior, D.; Stephan, H.; Dönker, N. Characterization of Etoposide- and Cisplatin-Chemoresistant Retinoblastoma Cell Lines. Oncol. Rep. 2018, 39, 160–172. [Google Scholar] [CrossRef]
  87. Khabipov, A.; Freund, E.; Liedtke, K.R.; Käding, A.; Riese, J.; van der Linde, J.; Kersting, S.; Partecke, L.I.; Bekeschus, S. Murine Macrophages Modulate Their Inflammatory Profile in Response to Gas Plasma-Inactivated Pancreatic Cancer Cells. Cancers 2021, 13, 2525. [Google Scholar] [CrossRef]
  88. Achkar, I.W.; Kader, S.; Dib, S.S.; Junejo, K.; Al-Bader, S.B.; Hayat, S.; Bhagwat, A.M.; Rousset, X.; Wang, Y.; Viallet, J.; et al. Metabolic Signatures of Tumor Responses to Doxorubicin Elucidated by Metabolic Profiling in Ovo. Metabolites 2020, 10, 268. [Google Scholar] [CrossRef] [PubMed]
  89. Winter, G.; Koch, A.B.F.; Löffler, J.; Jelezko, F.; Lindén, M.; Li, H.; Abaei, A.; Zuo, Z.; Beer, A.J.; Rasche, V. In Vivo PET/MRI Imaging of the Chorioallantoic Membrane. Front. Phys. 2020, 8, 151. [Google Scholar] [CrossRef]
  90. Miura, K.; Koyanagi-Aoi, M.; Maniwa, Y.; Aoi, T. Chorioallantoic Membrane Assay Revealed the Role of TIPARP (2,3,7,8-Tetrachlorodibenzo-p-Dioxin-Inducible Poly (ADP-Ribose) Polymerase) in Lung Adenocarcinoma-Induced Angiogenesis. Cancer Cell Int. 2023, 23, 34. [Google Scholar] [CrossRef] [PubMed]
  91. Ribatti, D. The Chick Embryo Chorioallantoic Membrane as an Experimental Model to Study In Vivo Angiogenesis in Glioblastoma Multiforme. Brain Res. Bull. 2022, 182, 26–29. [Google Scholar] [CrossRef]
  92. Damanskienė, E.; Balnytė, I.; Valančiūtė, A.; Alonso, M.M.; Preikšaitis, A.; Stakišaitis, D. The Different Temozolomide Effects on Tumorigenesis Mechanisms of Pediatric Glioblastoma PBT24 and SF8628 Cell Tumor in CAM Model and on Cells In Vitro. Int. J. Mol. Sci. 2022, 23, 2001. [Google Scholar] [CrossRef]
  93. Kerkhoff, M.; Grunewald, S.; Schaefer, C.; Zöllner, S.K.; Plaumann, P.; Busch, M.; Dünker, N.; Ketzer, J.; Kersting, J.; Bauer, S.; et al. Evaluation of the Effect of Photodynamic Therapy on CAM-Grown Sarcomas. Bioengineering 2023, 10, 464. [Google Scholar] [CrossRef]
  94. Guder, W.K.; Hartmann, W.; Buhles, C.; Burdack, M.; Busch, M.; Dünker, N.; Hardes, J.; Dirksen, U.; Bauer, S.; Streitbürger, A. 5-ALA-Mediated Fluorescence of Musculoskeletal Tumors in a Chick Chorio-Allantoic Membrane Model: Preclinical In Vivo Qualification Analysis as a Fluorescence-Guided Surgery Agent in Orthopedic Oncology. J. Orthop. Surg. Res. 2022, 17, 34. [Google Scholar] [CrossRef]
  95. Hu, L.; Li, K.; Lin, L.; Qian, F.; Li, P.; Zhu, L.; Cai, H.; You, L.; Song, J.; Kok, S.H.L.; et al. Reversine Suppresses Osteosarcoma Cell Growth through Targeting BMP-Smad1/5/8-Mediated Angiogenesis. Microvasc. Res. 2021, 135, 104136. [Google Scholar] [CrossRef]
  96. Fialho, S.L.; Silvestrini, B.R.; Vieira, J.; Paiva, M.R.B.; Silva, L.M.; Chahud, F.; Silva-Cunha, A.; Correa, Z.M.; Jorge, R. Successful Growth of Fresh Retinoblastoma Cells in Chorioallantoic Membrane. Int. J. Retina Vitreous 2020, 6, 33. [Google Scholar] [CrossRef]
  97. Merlos Rodrigo, M.A.; Casar, B.; Michalkova, H.; Jimenez Jimenez, A.M.; Heger, Z.; Adam, V. Extending the Applicability of In Ovo and Ex Ovo Chicken Chorioallantoic Membrane Assays to Study Cytostatic Activity in Neuroblastoma Cells. Front. Oncol. 2021, 11, 707366. [Google Scholar] [CrossRef] [PubMed]
  98. Barnett, S.E.; Herrmann, A.; Shaw, L.; Gash, E.N.; Poptani, H.; Sacco, J.J.; Coulson, J.M. The Chick Embryo Xenograft Model for Malignant Pleural Mesothelioma: A Cost and Time Efficient 3Rs Model for Drug Target Evaluation. Cancers 2022, 14, 5836. [Google Scholar] [CrossRef] [PubMed]
  99. Vu, B.T.; Shahin, S.A.; Croissant, J.; Fatieiev, Y.; Matsumoto, K.; Le-Hoang Doan, T.; Yik, T.; Simargi, S.; Conteras, A.; Ratliff, L.; et al. Chick Chorioallantoic Membrane Assay as an In Vivo Model to Study the Effect of Nanoparticle-Based Anticancer Drugs in Ovarian Cancer. Sci. Rep. 2018, 8, 8524. [Google Scholar] [CrossRef]
  100. Schneider-Stock, R.; Flügen, G. Editorial for Special Issue: The Chorioallantoic Membrane (CAM) Model—Traditional and State-of-the Art Applications: The 1st International CAM Conference. Cancers 2023, 15, 772. [Google Scholar] [CrossRef] [PubMed]
  101. Mangieri, D.; Nico, B.; Benagiano, V.; De Giorgis, M.; Vacca, A.; Ribatti, D. Angiogenic Activity of Multiple Myeloma Endothelial Cells In Vivo in the Chick Embryo Chorioallantoic Membrane Assayis Associated to a Down-Regulation in the Expression of Endogenous Endostatin. J. Cell. Mol. Med. 2008, 12, 1023–1028. [Google Scholar] [CrossRef] [PubMed]
  102. Weis, S.M.; Cheresh, D.A. Tumor Angiogenesis: Molecular Pathways and Therapeutic Targets. Nat. Med. 2011, 17, 1359–1370. [Google Scholar] [CrossRef]
  103. Hanahan, D.; Weinberg, R.A. The Hallmarks of Cancer Review Evolve Progressively from Normalcy via a Series of Pre. Cell 2000, 100, 57–70. [Google Scholar] [CrossRef]
  104. Folkman, J. Tumor Angiogenesis: Therapeutic Implications. N. Engl. J. Med. 1971, 285, 1182–1186. [Google Scholar] [CrossRef]
  105. Demcisakova, Z.; Luptakova, L.; Tirpakova, Z.; Kvasilova, A.; Medvecky, L.; De Spiegelaere, W.; Petrovova, E. Evaluation of Angiogenesis in an Acellular Porous Biomaterial Based on Polyhydroxybutyrate and Chitosan Using the Chicken Ex Ovo Chorioallantoic Membrane Model. Cancers 2022, 14, 4194. [Google Scholar] [CrossRef]
  106. Jilani, S.M.; Murphy, T.J.; Thai, S.N.M.; Eichmann, A.; Alva, J.A.; Luisa Iruela-Arispe, M. Selective Binding of Lectins to Embryonic Chicken Vasculature. J. Histochem. Cytochem. 2003, 51, 597–604. [Google Scholar] [CrossRef]
  107. Hagedorn, M.; Balke, M.; Schmidt, A.; Bloch, W.; Kurz, H.; Javerzat, S.; Rousseau, B.; Wilting, J.; Bikfalvi, A. VEGF Coordinates Interaction of Pericytes and Endothelial Cells During Vasculogenesis and Experimental Angiogenesis. Dev. Dyn. 2004, 230, 23–33. [Google Scholar] [CrossRef]
  108. Marinaccio, C.; Nico, B.; Ribatti, D. Differential Expression of Angiogenic and Anti-Angiogenic Molecules in the Chick Embryo Chorioallantoic Membrane and Selected Organs during Embryonic Development. Int. J. Dev. Biol. 2013, 57, 907–916. [Google Scholar] [CrossRef] [PubMed]
  109. Ribatti, D.; Alessandri, G.; Baronio, M.; Raffaghello, L.; Cosimo, E.; Marimpietri, D.; Montaldo, P.G.; De Falco, G.; Caruso, A.; Vacca, A.; et al. Inhibition of Neuroblastoma-Induced Angiogenesis by Fenretinide. Int. J. Cancer 2001, 94, 314–321. [Google Scholar] [CrossRef] [PubMed]
  110. Javanmardi, S.; Aghamaali, M.; Abolmaali, S.; Mohammadi, S.; Tamaddon, A.M. MiR-21, An Oncogenic Target MiRNA for Cancer Therapy: Molecular Mechanisms and Recent Advancements in Chemo and Radio-Resistance. Curr. Gene Ther. 2017, 16, 375–389. [Google Scholar] [CrossRef] [PubMed]
  111. Vimalraj, S.; Subramanian, R.; Saravanan, S.; Arumugam, B.; Anuradha, D. MicroRNA-432-5p Regulates Sprouting and Intussusceptive Angiogenesis in Osteosarcoma Microenvironment by Targeting PDGFB. Lab. Investig. 2021, 101, 1011–1025. [Google Scholar] [CrossRef]
  112. Ganesh, S.; Iyer, A.K.; Weiler, J.; Morrissey, D.V.; Amiji, M.M. Combination of SiRNA-Directed Gene Silencing with Cisplatin Reverses Drug Resistance in Human Non-Small Cell Lung Cancer. Mol. Ther. Nucleic Acids 2013, 2, e110. [Google Scholar] [CrossRef]
  113. Chan, J.K.; Blansit, K.; Kiet, T.; Sherman, A.; Wong, G.; Earle, C.; Bourguignon, L.Y.W. The Inhibition of MiR-21 Promotes Apoptosis and Chemosensitivity in Ovarian Cancer. Gynecol. Oncol. 2014, 132, 739–744. [Google Scholar] [CrossRef]
  114. Javanmardi, S.; Abolmaali, S.S.; Mehrabanpour, M.J.; Aghamaali, M.R.; Tamaddon, A.M. PEGylated Nanohydrogels Delivering Anti-MicroRNA-21 Suppress Ovarian Tumor-Associated Angiogenesis in Matrigel and Chicken Chorioallantoic Membrane Models. BioImpacts 2022, 12, 449–461. [Google Scholar] [CrossRef]
  115. Liu, Y.; Luo, F.; Wang, B.; Li, H.; Xu, Y.; Liu, X.; Shi, L.; Lu, X.; Xu, W.; Lu, L.; et al. STAT3-Regulated Exosomal MiR-21 Promotes Angiogenesis and Is Involved in Neoplastic Processes of Transformed Human Bronchial Epithelial Cells. Cancer Lett. 2016, 370, 125–135. [Google Scholar] [CrossRef]
  116. Tome, Y.; Kimura, H.; Kiyuna, T.; Sugimoto, N.; Tsuchiya, H.; Kanaya, F.; Bouvet, M.; Hoffman, R.M. Disintegrin Targeting of an α v β 3 Integrin-over-Expressing High-Metastatic Human Osteosarcoma with Echistatin Inhibits Cell Proliferation, Migration, Invasion and Adhesion in Vitro. Oncotarget 2016, 7, 46315–46320. [Google Scholar] [CrossRef]
  117. Maacha, S.; Saule, S. Evaluation of Tumor Cell Invasiveness in Vivo: The Chick Chorioallantoic Membrane Assay. In Methods in Molecular Biology—Chapter 8; Humana Press Inc.: Totowa, NJ, USA, 2018; Volume 1749, pp. 71–77. [Google Scholar]
  118. Shioda, T.; Munn, L.L.; Fenner, M.H.; Jain, R.K.; Isselbacher, K.J. Early Events of Metastasis in the Microcirculation Involve Changes in Gene Expression of Cancer Cells Tracking MRNA Levels of Metastasizing Cancer Cells in the Chick Embryo Chorioallantoic Membrane. Am. J. Pathol. 1997, 150, 2099. [Google Scholar]
  119. Cecilia Subauste, M.; Kupriyanova, T.A.; Conn, E.M.; Ardi, V.C.; Quigley, J.P.; Deryugina, E.I. Evaluation of Metastatic and Angiogenic Potentials of Human Colon Carcinoma Cells in Chick Embryo Model Systems. Clin. Exp. Metastasis 2009, 26, 1033–1047. [Google Scholar] [CrossRef]
  120. Deryugina, E.I.; Zijlstra, A.; Partridge, J.J.; Kupriyanova, T.A.; Madsen, M.A.; Papagiannakopoulos, T.; Quigley, J.P. Unexpected Effect of Matrix Metalloproteinase Down-Regulation on Vascular Intravasation and Metastasis of Human Fibrosarcoma Cells Selected In Vivo for High Rates of Dissemination. Cancer Res. 2005, 65, 10959–10969. [Google Scholar] [CrossRef]
  121. Deryugina, E.I.; Quigley, J.P. Chick Embryo Chorioallantoic Membrane Model Systems to Study and Visualize Human Tumor Cell Metastasis. Histochem. Cell Biol. 2008, 130, 1119–1130. [Google Scholar] [CrossRef]
  122. Mira, E.; Ana Lacalle, R.; Gómez-Moutón, C.; Leonardo, E.; Mañes, S. Quantitative Determination of Tumor Cell Intravasation in a Real-Time Polymerase Chain Reaction-Based Assay. Clin. Exp. Metastasis 2002, 19, 313–318. [Google Scholar] [CrossRef]
  123. Zijlstra, A.; Mellor, R.; Panzarella, G.; Aimes, R.; Hooper, J.; Marchenko, N.; Quigley, J. A Quantitative Analysis of Rate-Limiting Steps in the Metastatic Cascade Using Human-Specific Real-Time Polymerase Chain Reaction. Cancer Res. 2002, 62, 7083–7092. [Google Scholar]
  124. Augustine, R.; Alhussain, H.; Hasan, A.; Ahmed, M.B.; Yalcin, H.C.; Al Moustafa, A.E. A Novel in Ovo Model to Study Cancer Metastasis Using Chicken Embryos and GFP Expressing Cancer Cells. Bosn J. Basic Med. Sci. 2020, 20, 140–148. [Google Scholar] [CrossRef]
  125. Schneider, T.; Osl, F.; Friess, T.; Stockinger, H.; Scheuer, W. V Quantification of Human Alu Sequences by Real-Time PCR-an Improved Method to Measure Therapeutic Efficacy of Anti-Metastatic Drugs in Human Xenotransplants. Clin. Exp. Metastasis 2002, 19, 571–582. [Google Scholar] [CrossRef]
  126. Kim, J.; Yu, W.; Kovalski, K.; Ossowski, L. Requirement for Specific Proteases in Cancer Cell Intravasation as Revealed by a Novel Semiquantitative PCR-Based Assay. Cell 1998, 94, 353–362. [Google Scholar] [CrossRef]
  127. Kobayashi, T.; Koshida, K.; Endo, Y.; Imao, T.; Uchibayashi, T.; Sasaki, T.; Namiki, M. Basic Science A Chick Embryo Model for Metastatic Human Prostate Cancer. Eur. Urol. 1998, 34, 154–160. [Google Scholar] [CrossRef]
  128. Komatsu, A.; Matsumoto, K.; Saito, T.; Muto, M.; Tamanoi, F. Patient Derived Chicken Egg Tumor Model (PDcE Model): Current Status and Critical Issues. Cells 2019, 8, 440. [Google Scholar] [CrossRef]
  129. Marcion, G.; Hermetet, F.; Neiers, F.; Uyanik, B.; Dondaine, L.; Dias, A.M.M.; Da Costa, L.; Moreau, M.; Bellaye, P.S.; Collin, B.; et al. Nanofitins Targeting Heat Shock Protein 110: An Innovative Immunotherapeutic Modality in Cancer. Int. J. Cancer 2021, 148, 3019–3031. [Google Scholar] [CrossRef]
  130. Skowron, M.A.; Sathe, A.; Romano, A.; Hoffmann, M.J.; Schulz, W.A.; van Koeveringe, G.A.; Albers, P.; Nawroth, R.; Niegisch, G. Applying the Chicken Embryo Chorioallantoic Membrane Assay to Study Treatment Approaches in Urothelial Carcinoma. Urol. Oncol. Semin. Orig. Investig. 2017, 35, 544.e11–544.e23. [Google Scholar] [CrossRef] [PubMed]
  131. Swadi, R.; Mather, G.; Pizer, B.L.; Losty, P.D.; See, V.; Moss, D. Optimising the Chick Chorioallantoic Membrane Xenograft Model of Neuroblastoma for Drug Delivery. BMC Cancer 2018, 18, 28. [Google Scholar] [CrossRef]
  132. Eckrich, J.; Kugler, P.; Buhr, C.R.; Ernst, B.P.; Mendler, S.; Baumgart, J.; Brieger, J.; Wiesmann, N. Monitoring of Tumor Growth and Vascularization with Repetitive Ultrasonography in the Chicken Chorioallantoic-Membrane-Assay. Sci. Rep. 2020, 10, 18585. [Google Scholar] [CrossRef]
  133. Gilson, P.; Couvet, M.; Vanwonterghem, L.; Henry, M.; Vollaire, J.; Baulin, V.; Werner, M.; Orlowska, A.; Josserand, V.; Mahuteau-Betzer, F. The Pyrrolopyrimidine Colchicine-Binding Site Agent PP-13 Reduces the Metastatic Dissemination of Invasive Cancer Cells In Vitro and In Vivo. Biochem. Pharmacol. 2019, 160, 1–13. [Google Scholar] [CrossRef]
  134. Kleibeuker, E.A.; ten Hooven, M.A.; Castricum, K.C.; Honeywell, R.; Griffioen, A.W.; Verheul, H.M.; Slotman, B.J.; Thijssen, V.L. Optimal Treatment Scheduling of Ionizing Radiation and Sunitinib Improves the Antitumor Activity and Allows Dose Reduction. Cancer Med. 2015, 4, 1003–1015. [Google Scholar] [CrossRef]
  135. Marimpietri, D.; Brignole, C.; Nico, B.; Pastorino, F.; Pezzolo, A.; Piccardi, F.; Cilli, M.; Di Paolo, D.; Pagnan, G.; Longo, L.; et al. Combined Therapeutic Effects of Vinblastine and Rapamycin on Human Neuroblastoma Growth, Apoptosis, and Angiogenesis. Clin. Cancer Res. 2007, 13, 3977–3988. [Google Scholar] [CrossRef]
  136. Marimpietri, D.; Nico, B.; Vacca, A.; Mangieri, D.; Catarsi, P.; Ponzoni, M.; Ribatti, D. Synergistic Inhibition of Human Neuroblastoma-Related Angiogenesis by Vinblastine and Rapamycin. Oncogene 2005, 24, 6785–6795. [Google Scholar] [CrossRef]
  137. Ademii, H.; Shinde, D.A.; Gassmann, M.; Gerst, D.; Chaachouay, H.; Vogel, J.; Gorr, T.A. Targeting Neovascularization and Respiration of Tumor Grafts Grown on Chick Embryo Chorioallantoic Membranes. PLoS ONE 2021, 16, e0251765. [Google Scholar] [CrossRef]
  138. Katrancioglu, N.; Karahan, O.; Kilic, A.T.; Altun, A.; Katrancioglu, O.; Polat, Z.A. The Antiangiogenic Effects of Levosimendan in a CAM Assay. Microvasc. Res. 2012, 83, 263–266. [Google Scholar] [CrossRef]
  139. Khademhosseini, A.; Langer, R. A Decade of Progress in Tissue Engineering. Nat. Protoc. 2016, 11, 1775–1781. [Google Scholar] [CrossRef]
  140. Chocholata, P.; Kulda, V.; Babuska, V. Fabrication of Scaffolds for Bone-Tissue Regeneration. Materials 2019, 12, 568. [Google Scholar] [CrossRef]
  141. Donderwinkel, I.; Tuan, R.S.; Cameron, N.R.; Frith, J.E. Tendon Tissue Engineering: Current Progress towards an Optimized Tenogenic Differentiation Protocol for Human Stem Cells. Acta Biomater. 2022, 145, 25–42. [Google Scholar] [CrossRef]
  142. Gao, J.; Yu, X.; Wang, X.; He, Y.; Ding, J. Biomaterial–Related Cell Microenvironment in Tissue Engineering and Regenerative Medicine. Engineering 2022, 13, 31–45. [Google Scholar] [CrossRef]
  143. Wang, J.; Huang, D.; Yu, H.; Cheng, Y.; Ren, H.; Zhao, Y. Developing Tissue Engineering Strategies for Liver Regeneration. Eng. Regen. 2022, 3, 80–91. [Google Scholar] [CrossRef]
  144. Sainsbury, E.; Amaral, R.d.; Blayney, A.W.; Walsh, R.M.C.; O’Brien, F.J.; O’Leary, C. Tissue Engineering and Regenerative Medicine Strategies for the Repair of Tympanic Membrane Perforations. Biomater. Biosyst. 2022, 6, 100046. [Google Scholar] [CrossRef]
  145. Cao, S.; Zhao, Y.; Hu, Y.; Zou, L.; Chen, J. New Perspectives: In-Situ Tissue Engineering for Bone Repair Scaffold. Compos. B Eng. 2020, 202, 108445. [Google Scholar] [CrossRef]
  146. Arjunan, A.; Baroutaji, A.; Robinson, J.; Wang, C. Tissue Engineering Concept. In Encyclopedia of Smart Materials; Olabi, A.-G., Ed.; Elsevier: Oxford, UK, 2022; pp. 103–112. ISBN 978-0-12-815733-6. [Google Scholar]
  147. Blume, C.; Kraus, X.; Heene, S.; Loewner, S.; Stanislawski, N.; Cholewa, F.; Blume, H. Vascular Implants—New Aspects for in Situ Tissue Engineering. Eng. Life Sci. 2022, 22, 344–360. [Google Scholar] [CrossRef]
  148. Ding, T.; Kang, W.; Li, J.; Yu, L.; Ge, S. An In Situ Tissue Engineering Scaffold with Growth Factors Combining Angiogenesis and Osteoimmunomodulatory Functions for Advanced Periodontal Bone Regeneration. J. Nanobiotechnol. 2021, 19, 247. [Google Scholar] [CrossRef]
  149. Fu, L.; Li, P.; Li, H.; Gao, C.; Yang, Z.; Zhao, T.; Chen, W.; Liao, Z.; Peng, Y.; Cao, F.; et al. The Application of Bioreactors for Cartilage Tissue Engineering: Advances, Limitations, and Future Perspectives. Stem Cells Int. 2021, 2021, 6621806. [Google Scholar] [CrossRef]
  150. Radisic, M.; Marsano, A.; Maidhof, R.; Wang, Y.; Vunjak-Novakovic, G. Cardiac Tissue Engineering Using Perfusion Bioreactor Systems. Nat. Protoc. 2008, 3, 719–738. [Google Scholar] [CrossRef]
  151. Todros, S.; Spadoni, S.; Maghin, E.; Piccoli, M.; Pavan, P.G. A Novel Bioreactor for the Mechanical Stimulation of Clinically Relevant Scaffolds for Muscle Tissue Engineering Purposes. Processes 2021, 9, 474. [Google Scholar] [CrossRef]
  152. Montorsi, M.; Genchi, G.G.; De Pasquale, D.; De Simoni, G.; Sinibaldi, E.; Ciofani, G. Design, Fabrication, and Characterization of a Multimodal Reconfigurable Bioreactor for Bone Tissue Engineering. Biotechnol. Bioeng. 2022, 119, 1965–1979. [Google Scholar] [CrossRef]
  153. Sun, T.; Meng, C.; Ding, Q.; Yu, K.; Zhang, X.; Zhang, W.; Tian, W.; Zhang, Q.; Guo, X.; Wu, B.; et al. In Situ Bone Regeneration with Sequential Delivery of Aptamer and BMP2 from an ECM-Based Scaffold Fabricated by Cryogenic Free-Form Extrusion. Bioact. Mater. 2021, 6, 4163–4175. [Google Scholar] [CrossRef]
  154. Poudel, B.K.; Robert, M.C.; Simpson, F.C.; Malhotra, K.; Jacques, L.; Labarre, P.; Griffith, M. In Situ Tissue Regeneration in the Cornea from Bench to Bedside. Cells Tissues Organs 2021, 211, 506–526. [Google Scholar] [CrossRef]
  155. Periayah, M.H.; Halim, A.S.; Saad, A.Z.M. Chitosan: A Promising Marine Polysaccharide for Biomedical Research. Pharmacogn. Rev. 2016, 10, 39–42. [Google Scholar] [CrossRef]
  156. Pavlovic, M. Bioengineering—A Conceptual Approach; Springer: Berlin/Heidelberg, Germany; Florida Atlantic University: Boca Raton, FL, USA, 2015. [Google Scholar]
  157. Kohli, N.; Sharma, V.; Orera, A.; Sawadkar, P.; Owji, N.; Frost, O.G.; Bailey, R.J.; Snow, M.; Knowles, J.C.; Blunn, G.W.; et al. Pro-Angiogenic and Osteogenic Composite Scaffolds of Fibrin, Alginate and Calcium Phosphate for Bone Tissue Engineering. J. Tissue Eng. 2021, 12, 20417314211005610. [Google Scholar] [CrossRef]
  158. Eldeeb, A.E.; Salah, S.; Elkasabgy, N.A. Biomaterials for Tissue Engineering Applications and Current Updates in the Field: A Comprehensive Review. AAPS PharmSciTech 2022, 23, 267. [Google Scholar] [CrossRef]
  159. AL-Hamoudi, F.; Rehman, H.U.; Almoshawah, Y.A.; Talari, A.C.S.; Chaudhry, A.A.; Reilly, G.C.; Rehman, I.U. Bioactive Composite for Orbital Floor Repair and Regeneration. Int. J. Mol. Sci. 2022, 23, 333. [Google Scholar] [CrossRef]
  160. Ribatti, D.; Annese, T.; Tamma, R. The Use of the Chick Embryo CAM Assay in the Study of Angiogenic Activiy of Biomaterials. Microvasc. Res. 2020, 131, 104026. [Google Scholar] [CrossRef]
  161. Mishra, R.; Roux, B.M.; Posukonis, M.; Bodamer, E.; Brey, E.M.; Fisher, J.P.; Dean, D. Effect of Prevascularization on In Vivo Vascularization of Poly(Propylene Fumarate)/Fibrin Scaffolds. Biomaterials 2016, 77, 255–266. [Google Scholar] [CrossRef] [PubMed]
  162. Ignjatovic, N.; Ajdukovic, Z.; Uskokovic, D. New Biocomposite [Biphasic Calcium Phosphate/Poly-DL-Lactide-Co-Glycolide/Biostimulative Agent] Filler for Reconstruction of Bone Tissue Changed by Osteoporosis. J. Mater. Sci. Mater. Med. 2005, 16, 621–626. [Google Scholar] [CrossRef]
  163. Wittmann, K.; Storck, K.; Muhr, C.; Mayer, H.; Regn, S.; Staudenmaier, R.; Wiese, H.; Maier, G.; Bauer-Kreisel, P.; Blunk, T. Development of Volume-Stable Adipose Tissue Constructs Using Polycaprolactone-Based Polyurethane Scaffolds and Fibrin Hydrogels. J. Tissue Eng. Regen. Med. 2016, 10, E409–E418. [Google Scholar] [CrossRef]
  164. Schagemann, J.C.; Chung, H.W.; Mrosek, E.H.; Stone, J.J.; Fitzsimmons, J.S.; O’Driscoll, S.W.; Reinholz, G.G. Poly-ε-Caprolactone/Gel Hybrid Scaffolds for Cartilage Tissue Engineering. J. Biomed. Mater. Res. A 2010, 93, 454–463. [Google Scholar] [CrossRef] [PubMed]
  165. Panadero, J.A.; Vikingsson, L.; Gomez Ribelles, J.L.; Sencadas, V.; Lanceros-Mendez, S. Fatigue Prediction in Fibrin Poly-ε-Caprolactone Macroporous Scaffolds. J. Mech. Behav. Biomed. Mater. 2013, 28, 55–61. [Google Scholar] [CrossRef]
  166. Fu, S.Z.; Ni, P.Y.; Wang, B.Y.; Chu, B.Y.; Zheng, L.; Luo, F.; Luo, J.C.; Qian, Z.Y. Injectable and Thermo-Sensitive PEG-PCL-PEG Copolymer/Collagen/n-HA Hydrogel Composite for Guided Bone Regeneration. Biomaterials 2012, 33, 4801–4809. [Google Scholar] [CrossRef]
  167. Lee, K.Y.; Mooney, D.J. Hydrogels for Tissue Engineering. Chem. Rev. 2001, 101, 1869–1879. [Google Scholar] [CrossRef]
  168. Kocak, F.Z.; Talari, A.C.S.; Yar, M.; Rehman, I.U. In-Situ Forming Ph and Thermosensitive Injectable Hydrogels to Stimulate Angiogenesis: Potential Candidates for Fast Bone Regeneration Applications. Int. J. Mol. Sci. 2020, 21, 1633. [Google Scholar] [CrossRef]
  169. Conde-González, A.; Glinka, M.; Dutta, D.; Wallace, R.; Callanan, A.; Oreffo, R.O.C.; Bradley, M. Rapid Fabrication and Screening of Tailored Functional 3D Biomaterials: Validation in Bone Tissue Repair—Part II. Biomater. Adv. 2023, 145, 213250. [Google Scholar] [CrossRef] [PubMed]
  170. Okesola, B.O.; Mendoza-Martinez, A.K.; Cidonio, G.; Derkus, B.; Boccorh, D.K.; Osuna De La Peña, D.; Elsharkawy, S.; Wu, Y.; Dawson, J.I.; Wark, A.W.; et al. De Novo Design of Functional Coassembling Organic-Inorganic Hydrogels for Hierarchical Mineralization and Neovascularization. ACS Nano 2021, 15, 11202–11217. [Google Scholar] [CrossRef] [PubMed]
  171. Müller-Heupt, L.K.; Wiesmann-Imilowski, N.; Schröder, S.; Groß, J.; Ziskoven, P.C.; Bani, P.; Kämmerer, P.W.; Schiegnitz, E.; Eckelt, A.; Eckelt, J.; et al. Oxygen-Releasing Hyaluronic Acid-Based Dispersion with Controlled Oxygen Delivery for Enhanced Periodontal Tissue Engineering. Int. J. Mol. Sci. 2023, 24, 5936. [Google Scholar] [CrossRef] [PubMed]
  172. Mesa, F.L.; Aneiros, J.; Cabrera, A.; Bravo, M.; Caballero, T.; Revelles, F.; Del Moral, R.G.; O’Valle, F.J. Antiproliferative Effect of Topic Hyaluronic Acid Gel. Study in Gingival Biopsies of Patients with Periodontal Disease. Histol. Histopathol. 2002, 17, 747–753. [Google Scholar] [CrossRef] [PubMed]
  173. Eick, S.; Renatus, A.; Heinicke, M.; Pfister, W.; Stratul, S.-I.; Jentsch, H. Hyaluronic Acid as an Adjunct After Scaling and Root Planing: A Prospective Randomized Clinical Trial. J. Periodontol. 2013, 84, 941–949. [Google Scholar] [CrossRef] [PubMed]
  174. Decker, S.; Arango-Ospina, M.; Rehder, F.; Moghaddam, A.; Simon, R.; Merle, C.; Renkawitz, T.; Boccaccini, A.R.; Westhauser, F. In Vitro and in Ovo Impact of the Ionic Dissolution Products of Boron-Doped Bioactive Silicate Glasses on Cell Viability, Osteogenesis and Angiogenesis. Sci. Rep. 2022, 12, 8510. [Google Scholar] [CrossRef] [PubMed]
  175. Zhu, Q.X.; Li, Y.M.; Han, D. Co-Substitution of Carbonate and Fluoride in Hydroxyapatite: Effect on Substitution Type and Content. Front. Mater. Sci. 2015, 9, 192–198. [Google Scholar] [CrossRef]
  176. Barry, A.B.; Zhuang, H.; Baig, A.A.; Higuchi, W.I. Effect of Fluoride Pretreatment on the Solubility of Synthetic Carbonated Apatite. Calcif. Tissue Int. 2003, 72, 236–242. [Google Scholar] [CrossRef]
  177. Verma, G.; Barick, K.C.; Shetake, N.G.; Pandey, B.N.; Hassan, P.A. Citrate-Functionalized Hydroxyapatite Nanoparticles for PH-Responsive Drug Delivery. RSC Adv. 2016, 6, 77968–77976. [Google Scholar] [CrossRef]
  178. Khan, A.S.; Aamer, S.; Chaudhry, A.A.; Wong, F.S.L.; Rehman, I.U. Synthesis and Characterizations of a Fluoride-Releasing Dental Restorative Material. Mater. Sci. Eng. C 2013, 33, 3458–3464. [Google Scholar] [CrossRef]
  179. Da Mota, M.; De, V.; Branco, A. Polyurethane-Based Scaffolds for Bone Tissue Regeneration; Instituto Superior Técnico: Lisbon, Portugal, 2015. [Google Scholar]
  180. Bongio, M.; Lopa, S.; Gilardi, M.; Bersini, S.; Moretti, M. A 3D Vascularized Bone Remodeling Model Combining Osteoblasts and Osteoclasts in a CaP Nanoparticle-Enriched Matrix. Nanomedicine 2016, 11, 1073–1091. [Google Scholar] [CrossRef]
  181. Tadic, D.; Epple, M. A Thorough Physicochemical Characterisation of 14 Calcium Phosphate-Based Bone Substitution Materials in Comparison to Natural Bone. Biomaterials 2004, 25, 987–994. [Google Scholar] [CrossRef] [PubMed]
  182. Karageorgiou, V.; Kaplan, D. Porosity of 3D Biomaterial Scaffolds and Osteogenesis. Biomaterials 2005, 26, 5474–5491. [Google Scholar] [CrossRef] [PubMed]
  183. Vishnubalaji, R.; Atteya, M.; Al-Nbaheen, M.; Oreffo, R.O.C.; Aldahmash, A.; Alajez, N.M. Angio-genic Potential of Human Neonatal Foreskin Stromal Cells in the Chick Embryo Chorioallantoic Membrane Model. Stem Cells Int. 2015, 2015, 257019. [Google Scholar] [CrossRef]
  184. Black, C.; Kanczler, J.M.; de Andrés, M.C.; White, L.J.; Savi, F.M.; Bas, O.; Saifzadeh, S.; Henkel, J.; Zannettino, A.; Gronthos, S.; et al. Characterisation and Evaluation of the Regenerative Capacity of Stro-4+ Enriched Bone Marrow Mesenchymal Stromal Cells Using Bovine Extracellular Matrix Hydrogel and a Novel Biocompatible Melt Electro-Written Medical-Grade Polycaprolactone Scaf-fold. Biomaterials 2020, 247, 119998. [Google Scholar] [CrossRef] [PubMed]
  185. Saunders, M.M.; Simmerman, L.A.; Reed, G.L.; Sharkey, N.A.; Taylor, A.F. Biomimetic Bone Mechanotransduction Modeling in Neonatal Rat Femur Organ Cultures: Structural Verification of Proof of Concept. Biomech. Model. Mechanobiol. 2010, 9, 539–550. [Google Scholar] [CrossRef]
  186. Aldahmash, A.; Vishnubalaji, R. Transplantation of Human Neonatal Foreskin Stromal Cells in Ex Vivo Organotypic Cultures of Embryonic Chick Femurs. Saudi J. Biol. Sci. 2017, 24, 857–863. [Google Scholar] [CrossRef] [PubMed]
  187. Marshall, K.M.; Kanczler, J.M.; Oreffo, R.O.C. Evolving Applications of the Egg: Chorioallantoic Membrane Assay and Ex Vivo Organotypic Culture of Materials for Bone Tissue Engineering. J. Tissue Eng. 2020, 11, 2041731420942734. [Google Scholar] [CrossRef]
  188. Feder, A.-L.; Pion, E.; Troebs, J.; Lenze, U.; Prantl, L.; Htwe, M.M.; Phyo, A.; Haerteis, S.; Aung, T. Extended Analysis of Intratumoral Heterogeneity of Primary Osteosarcoma Tissue Using 3D- in-Vivo-Tumor-Model. Clin. Hemorheol. Microcirc. 2020, 76, 133–141. [Google Scholar] [CrossRef]
  189. Kanczler, J.M.; Smith, E.L.; Roberts, C.A.; Oreffo, R.O.C. A Novel Approach for Studying the Temporal Modulation of Embryonic Skeletal Development Using Organotypic Bone Cultures and Microcomputed Tomography. Tissue Eng. Part C Methods 2012, 18, 747–760. [Google Scholar] [CrossRef]
  190. Smith, E.; Kanczler, J.; Oreffo, R.O. A New Take on an Old Story: Chick Limb Organ Culture for Skeletal Niche Development and Regenerative Medicine Evaluation. Eur. Cell Mater. 2013, 11, 91–106. [Google Scholar] [CrossRef]
  191. Smith, E.L.; Rashidi, H.; Kanczler, J.M.; Shakesheff, K.M.; Oreffo, R.O.C. The Effects of 1α, 25-Dihydroxyvitamin D3 and Transforming Growth Factor-Β3 on Bone Development in an Ex Vivo Organotypic Culture System of Embryonic Chick Femora. PLoS ONE 2015, 10, e0121653. [Google Scholar] [CrossRef] [PubMed]
  192. Kaisto, S.; Saarela, U.; Dönges, L.; Raykhel, I.; Skovorodkin, I.; Vainio, S.J. Optimization of Renal Organoid and Organotypic Culture for Vascularization, Extended Development, and Improved Microscopy Imaging. J. Vis. Exp. 2020, 2020, e60995. [Google Scholar] [CrossRef]
Figure 1. (A) Observation of a chicken embryo at ED3 (scale bar = 1 mm). (B) Chicken embryo at ED3 in an ex ovo experiment (scale bar = 0.5 cm). (C) Hematoxylin and eosin staining of a chicken CAM at ED13; image taken using a Nikon microscope at 10× magnification. V: a major CAM blood vessel; arrow-head: sub-chorion capillaries.
Figure 1. (A) Observation of a chicken embryo at ED3 (scale bar = 1 mm). (B) Chicken embryo at ED3 in an ex ovo experiment (scale bar = 0.5 cm). (C) Hematoxylin and eosin staining of a chicken CAM at ED13; image taken using a Nikon microscope at 10× magnification. V: a major CAM blood vessel; arrow-head: sub-chorion capillaries.
Biology 12 01219 g001
Table 1. Tumor types tested in CAM in the last six years.
Table 1. Tumor types tested in CAM in the last six years.
OrgansResearch Papers
Prostate[81,82]
Colon[71,83]
Pancreas[84,85,86,87]
Breast[88,89]
Lung[90]
Glioblastoma[91,92]
Osteosarcoma[71,93,94,95]
Retinoblastoma[86,96]
Neuroblastoma[97]
Malignant Pleural Mesothelioma[98]
Ovary[99]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Palumbo, C.; Sisi, F.; Checchi, M. CAM Model: Intriguing Natural Bioreactor for Sustainable Research and Reliable/Versatile Testing. Biology 2023, 12, 1219. https://doi.org/10.3390/biology12091219

AMA Style

Palumbo C, Sisi F, Checchi M. CAM Model: Intriguing Natural Bioreactor for Sustainable Research and Reliable/Versatile Testing. Biology. 2023; 12(9):1219. https://doi.org/10.3390/biology12091219

Chicago/Turabian Style

Palumbo, Carla, Federica Sisi, and Marta Checchi. 2023. "CAM Model: Intriguing Natural Bioreactor for Sustainable Research and Reliable/Versatile Testing" Biology 12, no. 9: 1219. https://doi.org/10.3390/biology12091219

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop