Skip to main content

Molecular perspectives on systemic priming and concomitant immunity in colorectal carcinoma

Abstract

The progression of metastasis, a complex systemic disease, is facilitated by interactions between tumor cells and their isolated microenvironments. Over the past few decades, researchers have investigated the metastatic spread of cancer extensively, identifying multiple stages in the process, such as intravasation, extravasation, tumor latency, and the development of micrometastasis and macrometastasis. The premetastatic niche is established in target organs by the accumulation of aberrant immune cells and extracellular matrix proteins. The “seed and soil” idea, which has become widely known and accepted, is being used to this day to guide cancer studies. Changes in the local and systemic immune systems have a major impact on whether an infection spreads or not. The belief that the immune response may play a role in slowing tumor growth and may be beneficial against the metastatic disease underpins the responsiveness shown in the immunological landscape of metastasis. Various hypotheses on the phylogenesis of metastases have been proposed in the past. The primary tumor’s secreting factors shape the intratumoral microenvironment and the immune landscape, allowing this progress to be made. Therefore, it is evident that among disseminated tumor cells, there are distinct phenotypes that either carry budding for metastasis or have the ability to obtain this potential or in systemic priming through contact with substantial metastatic niches that have implications for medicinal chemistry. Concurrent immunity signals that the main tumor induces an immune response that may not be strong enough to eradicate the tumor. Immunotherapy’s success with some cancer patients shows that it is possible to effectively destroy even advanced-stage tumors by modifying the microenvironment and tumor-immune cell interactions. This review focuses on the metastasome in colorectal carcinoma and the therapeutic implications of site-specific metastasis, systemic priming, tumor spread, and the relationship between the immune system and metastasis.

Introduction

Cutting-edge technological developments and newer attractive models make a more accessible platform to study metastasis [1]. Approaches like single-cell sequencing of disseminated tumors are very much encouraging to learning metastasome and right opportunities to make some novel idea concerning ideas therapeutic potential [2]. Metastasome is responsible for adhesion, migration, and proliferation. It works as a mediator of intercellular communication [3]. It was a long back to find the history of malignant disease, and literature says it was found in Egypt in 1500 BC [4]. Hippocrates (fifth-century BC) was the first to see the scary lesions in mammary gland carcinoma patients [4]. Despite the several models to find the mechanism of tumor metastasis, it is mysterious and inconclusive clinically [5]. Metastasis is the leading cause of cancer morbidity and mortality, accounting for around 90% of cancer deaths [6]. Even though the cancer survival rate has been meaningfully improved over the years, progress is mainly due to initial diagnosis and malignancy growth inhibition [7]. Partial success has been made in considering cancer metastasis comprising colorectal carcinoma, though existing new-generation anticancer drugs do have effects on metastasis besides their special effects on cancer growth [8]. The cascade includes the development of angiogenesis, detachment, and migration of metastatic cells from the primary tumor, through the membrane and extracellular matrix (ECM) [9]. Then the process continues with the invasion of the membrane supporting endothelium of local blood in addition to lymphatic vessels and then also intravasation of metastatic cells. It is followed by the adhesion of circulating metastatic cells to the endothelium of capillaries of target organs and finally growth of secondary tumors at the target organ site [10]. Researchers have found that metastases, along with metastatically induced cancer cell clone, reveal definite features at the gene, protein, and epigenetic level and in functional level [11]. Physiologically, cells remain within their demarcated periphery through tight cell-cell adhesion and cell-matrix adhesion [10]. Metastatic lesions have less difference with primary tumors, but the acceleration from growth rate in dissemination condition was found in most of the cancers including colorectal carcinoma [12]. Cell adhesion is also important for cancer metastasis, and it is involved in settling metastatic cells at a distal site. It is well known that secondary lesions arise from the seeding and migration of primary tumor cells [13]. Newly, the interplay between primary tumor and secondary organs via circulation has transpired as a crucial factor of tumor metastasis [14]. Neoplastic cells can crosstalk with their surroundings by simple communications, whereas a primary tumor could possibly employ more refined mechanisms to accomplish efficient interactions with distant regions [15]. Antigens should be transported from the tumor site to a lymph node by migratory dendritic cells, and they left the tumor microenvironment or become dysfunctional. If tumor antigen is accepted as “self” during antigen presentation to CD4+T cells, expansion of immunosuppressive regulatory cells can be induced.

Seed and the soil theory

The “seed and soil” theory of metastasis puts out by Steven Paget was based on a review of 735 fatal breast cancer cases, each having an autopsy and several other cancer cases from the literature. In 1889, the Lancet published his research findings. The “seed” refers to specific tumor cells with the ability to metastasize, and the “soil” is any organ or tissue offering the ideal environment for the growth of the seeds, in his view, such that the distribution of metastases cannot be the result of chance [16]. James Ewing [17] disputed Paget’s “seed and soil” idea in 1928 and proposed that the anatomical makeup of the circulatory system results in pure mechanical processes that cause metastatic dispersion. The idea of “seed and soil” faded into obscurity for the following several decades as a result of not becoming the majority. Hart and Fidler’s groundbreaking from the 1980s confirmed Paget’s “seed and soil” idea by demonstrating the preferred homing of B16 melanoma tumor cells in particularly remote areas. Their research unequivocally established that despite all organs’ vasculatures containing potentially metastatic tumor cells, some organs experienced metastasis development [18].

Road to study metastasome: from metastasis to metastasis of metastasis

Tumor metastasis is a multistep and multistage process through which malignant cells extend from the primary tumor to intermittent organ sites. Even though tumor metastasis can happen initially in the tumor development period when the primary tumor is small or even undetectable, most happens later when the primary tumor is larger in shape [19]. Researchers have observed that overgrowth and supremacy within primary as well as secondary lesions by a solitary tumor cell population, with identical metastatic signature, are connected with the early metastasis phenomenon [20]. An extension of this hypothesis is that the late appearance of metastatic clones may result in different expression patterns between primary tumors and metastases, secondary to hiding the metastatic signatures in the primary tumors, by continuing non-metastatic clones [21]. On the other hand, investigation of clonal cell lines derived from late-stage carcinomas like colorectal carcinoma has provided direct evidence that discrete cancer cells were coexisting inside a tumor, contrasting in their respective metastatic competence, comprising ones that are non-metastatic, and confirming tumor heterogeneity shown in preclinical investigations with rodent model as well as human tumors [22].

In the year 1889, Stephen Paget, an English surgeon, proposed the “seed and soil” hypothesis. In recent years, more fundamental discoveries have conveyed understanding into our basic understanding of cancer metastasis, and numerous innovative concepts have been recognized [23]. For instance, the “tumor self-seeding hypothesis debated that circulating tumor cells might seed not only to local and distant organs in the body system but also to the original source i.e., primary tumor” [24]. Pre-metastatic niche, hypothesized as a fertile soil favorable to the existence and extension of metastatic seed, has concerned more attention in the period of metastasis research [25]. In past few decades, improvements in technologies like mass spectrometry, microarray, and genome sequencing knowledge have dramatically faster the attempt to widely illustrate the role of metastatic cancer cells and the relationship between primary and secondary tumors [26]. Especially, single-cell sequencing has appeared as a potent technology to characterize the landscape of individual cancer cells like colorectal carcinoma instead of investigating bulk tissue samples composed of millions of cells and has delivered novel insights into our understanding of the multifaceted metastasis systems [27]. Improvement of different microscopic techniques and imaging technology has empowered the visualization and analysis of neoplastic cell dynamics in experimental animals, which may consequently lead to innovative outcomes in metastasis research and promising therapeutic interventions [28].

The generic name for the process by which cancer cells move from the main tumor to nearby tissues and distant organs is metastasis, which accounts for the majority of cancer-related morbidity and mortality [29]. Many sequential and connected processes are involved in metastasis (Table 1). Colon cancer cells must separate from the primary tumor, intravagate to the circulatory and lymphatic systems, avoid the immune response, extravagate at distant capillary beds, and then penetrate and multiply in distant organs in order to complete the metastatic cascade.

Table 1 Origin of metastatic colorectal carcinoma cells

The metastatic niche and formation of macro-metastasis in colon cancer

The metastatic niche hypothesis states that while a cell’s precise genetic makeup unquestionably plays a key role in creating its malignant phenotype, microenvironmental conditions are also crucial in allowing malignant cells to reach their potential for metastatic spread. According to the metastatic niche concept, as a tumor moves from a micrometastatic to a macro metastatic state, an appropriately favorable milieu must develop for tumor cells to engraft and multiply at secondary sites [30]. Different models of metastasis phylogenesis have been studied earlier, ranging from the supposition of a common clonal origin for both primary tumor and metastases up to hypotheses of an entirely independent origin of metastasis as well as primary tumor [31]. Allgayer and his group (2020) established strong evidence for a common clonal ancestor of the primary tumor and corresponding metastasis [32]. The hypotheses on an entirely self-governing phylogenesis of a primary tumor and metastasis which is very unlikely at least for that tumor entity [32]. Klein and his group in 2009 worked on a mouse model, and they hypothesized that dissemination of metastatically proficient cells by primary tumor takes place in the initial stages of primary tumor development [33]. They concluded that primary and also secondary lesions of that cancer cells are developing separately.

The linear progression model (type of classical model) accepts metastasis as a chronological event resulting development of primary tumor in one direction [34]. In addition, in one study, Mantovani and his colleagues (2019) detected a tumor protein 53 (TP53) mutation in metastatic condition [35], whereas conforming primary tumor displayed a large deletion spanning TP53 regulator ATM serine/threonine kinase [36].

Since the 1990s, it is known that specific molecules that are detected on some of the disseminated tumor cells for instance urokinase-type plasminogen activator receptor (UPAR) in gastrointestinal carcinoma, epidermal growth factor receptor 2 (EGFR2) in mammary gland carcinoma, etc. associate with later clinical consequence, mainly tumor recurrence as well as metastasis [37]. Thus, it is clear that among disseminated tumor cells, particular phenotypes either already carry the potential for metastasis or have this during the dissemination process [38]. Another metastasis model (also called the self-seeding tumor model), where cancer cells are furthermore extended by metastasis and return to the primary tumor (Fig.1) to contribute to the dynamics of primary tumor genesis [39]. Separation of metastatically relevant clones from their common ancestor in the metastatic process may depend on the timing of specific epigenetic changes as to their consequence for metastatic capabilities [40]. This may be reflect a parallel progression model in cases of prior clonal separation or a linear progression model in cases of later clonal separation [41].

Fig. 1
figure 1

Metastasis of colorectal carcinoma

Allgayer and his colleagues (2020) analyzed genomic data of 12 colorectal paired metastasis/primary tumor cases, and they found strong evidence that metastasis every time harbors genomic changes that are private to metastasis and not shared with its analogous primary tumor and vice versa [32]. Generally, genomic lesions or metastasis-specific mutations that happened after clonal separation are supported by a single-cell sequencing study of Leung and his colleagues (2017) [42]. Vermaat and his colleagues (2012) conclude a late dissemination model based on their observations in patients with colorectal carcinoma with liver metastases [43]. Based on the literature for colorectal carcinoma, it can be suggested that a common ancestor clone between primary tumor and metastasis, along with initial heterogeneous dissemination that is independent, plays a role in clonal separation [44]. With the concept of metastasome, scientists have suggested two mechanistic models (Fig.2) for tumor metastasis [4], and they are as follows:

  1. a)

    Tumor organ training (TOTr) model: Researchers have found that exosomes can induce the expression of a set of metastatic genes in experimental mice models to assist cancer cell recruitment, trapping, and subsequently growth. Furthermore, subsets of metastasis endorsing microvesicles with CSCs property are capable of persuading pre-metastatic niche formation along with metastasis [4].

  2. b)

    Tumor organ targeting (TOTa) model: This model fundamentally looks similar to the genometastasis model that states about the cell-free DNAs originated from primary tumors. This cell-free DNA may release into the circulation of oncogenic patients and can be taken up by noncancerous cells in neighboring sites, leading to their transformation as well as the development of second primary tumors. Ghasemi and his groups (2013) proposed a TOTa model which rationalizes the organ tropism of tumor metastasis along with some explanations for morphological similarities between metastases and primary tumors [4].

Fig. 2
figure 2

Tumor organ training model and tumor organ targeting models of metastasome and the role of metastatic stem cells and cancer stem cells

Pathogenesis of cancer metastasis: link between concomitant immunity and metastatic cascade

The large list of consecutive, connected processes that make up the cancer-spreading process is extensive. Neoplasms experience a number of alterations over the course of the disease, according to clinical and experimental investigations. One of the major problems to find a therapy for most solid cancers, including colorectal carcinoma, is not the elimination of the primary tumor but rather the eradication of metastases [45]. Metastases upsurge from solitary tumors after neoplastic cells undergo distinctive alterations and headway over a multistep metastatic cascade, forming disseminated tumors which are tough to control. Tumor metastatic progression consists of (a) invasion of neoplastic cells into neighboring tissue at primary tumor site, (b) intravasation of metastatic cells into blood or lymph vessels, (c) survival in circulatory system, (d) extravasation from circulatory system to distant sites, and (e) proliferation in the new environment. Metastasis is a highly complex process [46], and in each step of metastatic cascade, immunogenic cancer cells may be recognized by the host immune system [47]. In addition, under certain conditions, researchers have found that some immune cells in fact favor metastatic disease along with tumor growth [48]. Experimental murine models of metastasis indicated that the advanced growth of a primary tumor inhibited the growth of an experimentally implanted secondary tumor via an immune mechanism, which is referred to as concomitant immunity (CI) (Fig 3) [49].

Fig. 3
figure 3

Concomitant immunity and tumor growth

Colorectal cancer stem cell and metastasome in immunosuppressive tumor microenvironment

Capability to generate new tumors may be found back to a few numbers of cells within a solid tumor. Cancer stem cell (CSC) hypothesis has been assumed to find the properties of this distinct subset of cells [50]. More investigations of their potential involvement in metastasis resulted in the introduction of a new term, namely metastatic stem cell (metSC) [51]. Remarkably, more investigations publicized that a specific genetic alteration is connected with an amplified expression of CSC-specific transcription factor resembling achaete-scute homolog 2 (ASCL2) in addition to insulin-like growth factor 2 (IGF2) and higher expression of SRY-box 9 (SOX9) and olfactomedin 4 (OLFM4) [52]. They are also identified to be associated in the presence of CSCs in tumors [53].

The presence of metastasis-specific genes or factors like forkhead box protein P2 (FOXP2), protein kinase B (PKB), BRCA2, and casitas B-lineage lymphoma (CBL) is responsible for stemness and self-renewal features of metastasis [54]. MicroRNAs (miRNAs) may play a vital role in the progress of CSCs. Overexpression of miRNA like miR-142-3p leads to loss of breast-CSCs properties supplemented by a reduced expression of CD44 and CD133 and a cell division protein BOD1 (biorientation of chromosomes in cell division protein 1), which are related to cellular stemness [55]. Notch (neurogenic locus notch homolog protein) signaling plays an important role in the existence of CSCs or MetSCs, and it was shown an uncertain performance in the development and progression of various cancers [56] including colorectal carcinoma. Molecular expressions at the functional level of the oncogenetic process will enhance a better idea and clinical relevance of metastasome.

Metastasis and intratumoral immune cell crosstalk in colorectal carcinoma

Several mechanisms are involved in making an immunosuppressed tumor microenvironment to enable immune escape, thereby motivating tumor progression as well as metastasis formation in colorectal carcinoma. Tumor-derived chemokine like CCL2 recruits monocytes into the tumor microenvironment, and these engaged monocytes can differentiate into tumor-associated macrophages [57]. Regulatory T cells can bind to cytokine IL-2 at a greater affinity than do effector T cells, starving CD8+T cells of IL-2 necessary for CTL activation [58]. Regulatory B cells can produce the inhibitory cytokines IL-10 that may inhibit effector functions of CTLs [59], and it can promote the conversion of CD4+ T cells into regulatory T cells [60]. These mechanisms may contribute to the possibility that tumors can escape immune recognition as well as destruction. CTLs are referred to as the foremost anticancer effector cells [61], and their priming by APCs is the starting phase for effective T-cell reactions. Thus, T-cell priming may be effective along with CTL responses in tumor cells.

Position-specific metastasis of colorectal carcinoma: ratification from metastasome

Ishaque and his colleagues (2018) have suggested that, during evolution of a metastatic clone that is proficient to colonize a specific organ, also programs for site-specific metastasis [36]. Researchers found a boosted mutational frequency inside PI3K-AKT axis in liver metastases and SRC/PI3K-AKT stimulation for colorectal-liver metastasis [62]. The mechanisms leading to SRC activity during the metastasis process are yet to be completely investigated [63]. In carcinomas, the importance of epithelial-to-mesenchymal transition (EMT) is well documented as an important mechanism to drive metastatic invasion [64]. Concisely, EMT involves in a process of loss of adherents and also tight junctions in epithelial cells which are described by downregulation of E-cadherin expression in addition to the loss in cell polarity [65]. The opposite process of mesenchymal-to-epithelial transition (MET) in contrast to epithelial EMT must be critical features of metSC to propagate and form colonies at target [66]. Ishaque and his group (2018) found that some miRNAs and their regulatory activity make a network of target specifically E-cadherin, SET domain containing 2 (SETD2), forkhead box N3 (FOXN3), zinc finger E-box-binding homeobox transcription factors (ZEB-TF), etc. that are involved in EMT/MET process and thus invasion as well as metastasis [36]. This opinion improves the scenario that EMT/MET organizing processes are crucial for metastasis of colorectal carcinoma to neighboring organs [67].

Vogelstein and his colleagues (1993) postulated a progression model of colorectal carcinoma suggesting consecutive mutational events in diverse signaling pathways [68]. Various studies have shown that alteration of signaling pathways like Wnt, TP53, and TGFβ is related to progression and metastasis of colorectal carcinoma [69]. A whole-genome study was done by Allgayer and his group (2020) to compare primary tumors with corresponding metastases form of colorectal carcinoma, and they confirmed the “Vogelstein sequence” [39]. Furthermore, interesting noncoding mutational events may enhance the progression model and ought to be explored functionally in future investigations [70]. Binnewies and his colleagues (2018) have found that metastatic progression, as well as metastatic switch, is associated with alterations of tumor immune microenvironment (TIME) performed in modulation of immune evasion [71].

Müller and his group (2001) have shown that chemokine induces rearrangement of cytoskeleton, adhesion of integrin, directional migration, etc. that are vital for localization of metastatic cancer cells to selective sites [72]. A number of researchers have reported the role of chemokine receptor CXCR4 and its ligand CXCL12 in metastasis [73] in many cancers including colorectal carcinoma.

Metastasome and its clinical implications

A recent whole-genome analysis study was conducted by Ishaque and his colleagues (2018) who found an indication that the metastases harbor genomic changes and are not being shared with the primary tumor [36]. Another methodological explanation may be that cancer cell population is giving rise to metastasis that is present in analogous primary tumor; however, in cells, numbers are too small to detect using the genetic signature of other subclones. This undoubtedly may be one clarification for an unaffected situation that metastases still arise afterward total therapeutic removal of primary tumor which causes maximum deaths related to cancer including colorectal carcinoma [74]. Different approaches of tumor cell migration that may permit therapeutic consequences at the individual level and initiatives must be propelled by combining imaging (micro with macroscopic level) synergistically [75].

Innovative molecular tracer’s techniques are used to visualize genomic changes in metastasis conditions, and this can be presented in the oncology department for the diagnostic purposes [76]. Recently, innovative biomarkers were found in primary tumors as well as in metastasis utilizing for a diagnosis like EGFR-based therapies, namely cetuximab [77]. They found that metastasis special mutations occur within FAT1, which modulates EMT and metastatic stemness and also induces unusual Wnt signaling during colorectal carcinoma, and these observations have the potential to make a strategy for targeted treatment [78]. Vermaat and his colleagues (2012) studied the role of mutated kinase insert domain receptor (KDR) and vascular endothelial growth factor receptor 1 (VEGFR1) gene which are related to colorectal carcinoma metastases and might have therapeutic potential [43]. Scientists have studied SMAD2 and SMAD4 proteins during colorectal carcinoma progression and genomic alterations within TGFβ pathway to find a new therapeutic strategy [79]. SRC inhibitors like dasatinib are now being used as a tool against liver metastases of colorectal carcinoma, after proper understanding of the role of SRC and its activation during metastasis of colorectal carcinoma [80].

Concomitant immunity as a therapeutic target to prevent metastasis

Concomitant immunity is the occurrence of secondary tumor rejection during primary tumor growth, seen in many experimental animal models of malignancy. It can be incited by numerous tumor-determined stimuli, and diverse immune cell subsets may either promote or hinder metastasis. Important actors are T cells, NK cells, and M1-like macrophages that can perceive and slaughter metastatic cancer cells in addition to Tregs and M2-like macrophages that are modified by the tumor to go around concomitant immunity over inhibition of T cells and NK cells. Different studies exhibit how inhibition of explicit concomitant immunity mechanisms quickens metastatic growth. Consequently, expanded comprehension of concomitant immunity may give a few new focuses to cancer treatment. Concomitant immunity appears to often weaken as time advances, and metastasis happens [49]. Hindrance of prostaglandin E2 synthesis reestablished the anti-metastatic impact of late concomitant immunity macrophages [49]. This model highlights the significance of new investigations, as they legitimately recommend explicit mediations to support concomitant immunity against metastasis. For instance, explicit hindrance or depletion of Tregs would fortify cytotoxic CD8+ T cell and NK cell function in both primary tumors and in circulation. This may prevent the early dissemination of malignancy cells from the primary tumor while expanding anti-tumor impact against already disseminated tumor cells in circulation or recently seeded tumor cells at other sites [81]. Remarkably, ongoing proof recommends that isoform-explicit hindrance of PI3K-Akt pathway specially inhibits Tregs with insignificant impact on concomitant immunity as a therapeutic target to forestall metastasis.

Metastasis as a systemic disease: molecular insights and clinical implications

The main factor contributing to cancer-related death is metastasis. Since primary tumors can change their local and systemic surroundings in ways that promote metastasis development, recent results from experimental models and clinical experience imply that cancer is a systemic disease even at early stages [82]. Even in individuals with early-stage disease, disseminated and circulating tumor cells can be found [83]; while not all of these cells progress into full-blown metastatic lesions, their presence signals metastatic spread and supports clinical prognostication. Numerous tumor-derived substances affect distant organs systemically and activate local inflammatory and mesenchymal cells to generate pre-metastatic and post-dissemination niches that promote metastatic expansion. In keeping with this, immunotherapy [84, 85] is one of the few curative treatments even for diffusely metastatic illness; however, it has only been successful in treating a small subset of patients and certain cancer types. The development of novel therapeutic strategies [86] focused on treating disseminated disease should be made easier with a better understanding of tumor-induced and host-related systemic implications on metastasis.

Conclusion and future aspect

Malignancy can be extended throughout the body of malignant patients via circulating tumor-derived microvesicles, but cell migration is a vital phenomenon that must be accomplished for fulfillment of the progression. Predominantly, the association and structure of cells basically malformed in the time of neoplastic development, and cancer is a disease of a precise tissue, not cell, thus gaining immense clinical relevance. An innovative concept like metastasome and its expanded information can generate novel concepts into the existing body of information to make a novel therapeutic approach in the field of oncology and will be a useful tool for clinicians in dealing with the scenario.

Upcoming therapies for metastatic colorectal carcinoma would perhaps be oriented to target the previously disseminated tumor cells in addition to damaging their growth and development into clinically relevant metastases since this is the controlling step in metastatization for most carcinomas. In this aspect, it will be vital to find an extensive genomic as well as molecular characterization of metastases related to their primary tumors’ active epigenetic drivers of metastasis. Metastasome is an emerging field with great promise, but until today, insufficient research work has been done using its noble concept. We hope the study of metastasome in addition to novel-targeted therapeutic approaches will be helpful to cancer patients in the modern era of precision medicine.

Not only primary tumor but also furthermore plastic nature of individual immune cells as well as functions can move the tumor immune microenvironment in the direction of an immunosuppressive, pro-tumor environment, debilitating concomitant immunity and empowering immune escape. Furthermore, the naturally happening metastatic process of tumor cell separation from the primary tumor, intravasation, endurance in circulation, and extravasation into target tissue are all not reiterated in models where a direct infusion of secondary tumor cell inoculum recreates metastasis. Taking everything into account, concomitant immunity plays a significant and varied role in all steps of the metastatic cascade. Numerous particular targets in the association cons associated with concomitant immunity and metastatic cascade have been recognized, taking into account the rational design of interventions that fortify the concomitant immunity to forestall metastasis and in this way diminish cancer morbidity as well as mortality.

Availability of data and materials

Data of the review are publicly published data. The study does not require ethical approval.

Abbreviations

BC:

Before Christ

ECM:

Extracellular matrix

TP53:

Tumor protein 53

ATM:

Ataxia-telangiectasia mutated

TOTr:

Tumor organ training

TOTa:

Tumor organ targeting

CSC:

Colorectal carcinoma

Metsc:

Metastatic stem cell

ASCL2:

Achaete-scute homolog 2

IGF2:

Insulin-like growth factor 2

SOX9:

SRY-box 9

OLFM4:

Olfactomedin 4

UTR:

Untranslated region

FOXP2:

Forkhead box protein P2

PKB:

Protein kinase B

BRCA:

BReastCAncer gene

CBL:

Casitas B-lineage lymphoma

MiRNAs:

MicroRNAs

BOD1:

Biorientation of chromosomes in cell division protein 1

NOTCH:

Neurogenic locus notch homolog protein

PI3K-AKT:

Phosphatidylinositol 3-kinase/protein kinase B

SRC:

SRC proto-oncogene, non-receptor tyrosine kinase

EMT:

Epithelial-to-mesenchymal transition

MET:

Mesenchymal-to-epithelial transition

ZEB-TF:

Zinc finger E-box-binding homeobox-transcription factors

FOXN3:

Forkhead box N3

SETD2:

SET domain containing 2

EGFR:

Epidermal growth factor receptor

FAT:

FAT atypical cadherin 1

KDR:

Kinase insert domain receptor

TGFβ:

Transforming growth factor beta

mTOR:

Mammalian target of rapamycin

References

  1. Cuadrado LG, Tracey N, Ma R, Qian B, Brunton VG. Mouse models of metastasis: progress and prospects. Disease Model Mechan. 2017;10:1061–74.

    Article  Google Scholar 

  2. Lawson DA, Kessenbrock K, Davis RT, Pervolarakis N, Werb Z. Tumour heterogeneity and metastasis at single-cell resolution. Nat Cell Biol. 2018;20(12):1349–60.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  3. Maia J, Caja S, Carolina M, Moraes S, Couto N, Costa-Silva B. Exosome-based cell-cell communication in the tumor microenvironment. Front Cell Dev Biol. 2018;6:18.

    Article  PubMed Central  PubMed  Google Scholar 

  4. Ghasemi R, Grassadonia A, Tinari N, Piccolo E, Natoli C, Tomao F, et al. Tumor-derived microvesicles: the metastasomes. Med Hypotheses. 2013;80:75–82.

    Article  CAS  PubMed  Google Scholar 

  5. Hunter KW, Crawford NPS, Alsarraj J. Mechanisms of metastasis. Breast Cancer Res. 2008;10(Suppl 1):S2.

    Article  PubMed Central  PubMed  Google Scholar 

  6. Seyfried TN, Huysentruyt LC. On the origin of cancer metastasis. Crit Rev Oncog. 2013;18(1-2):43–73.

    Article  PubMed Central  PubMed  Google Scholar 

  7. Guan X. Cancer metastases: challenges and opportunities. Acta Pharm Sin B. 2015;5(5):402–18.

    Article  PubMed Central  PubMed  Google Scholar 

  8. Seebacher NA, Stacy AE, Porter GM, et al. Clinical development of targeted and immune based anti-cancer therapies. J ExpClin Cancer Res. 2019;38:156. https://doi.org/10.1186/s13046-019-1094-2.

    Article  CAS  Google Scholar 

  9. Paolillo M, Schinelli S. Extracellular matrix alterations in metastatic processes Int. J Mol Sci. 2019;20:4947. https://doi.org/10.3390/ijms20194947.

    Article  CAS  Google Scholar 

  10. Ryen SVD, Deldicque L. The regulation of the metastatic cascade by physical activity: a narrative review. Cancers. 2020;12(1):153.

    Article  Google Scholar 

  11. Patel SA, Vanharanta S. Epigenetic determinants of metastasis. MolOncol. 2017;11(1):79–96.

    Google Scholar 

  12. Pantel K, Cote RJ, Fodstad O. Detection and clinical importance of micrometastatic disease. JNCI. 1999;91(13):1113–24.

    Article  CAS  PubMed  Google Scholar 

  13. Nguyen MJ. Genetic determinants of cancer metastasis. Nat Rev Genet. 2007;8:341–52.

    Article  CAS  PubMed  Google Scholar 

  14. Liu Q, Zhang H, Jiang X, Qian C, Liu Z, Luo D. Factors involved in cancer metastasis: a better understanding to “seed and soil” hypothesis. Mol Cancer. 2017;16:176.

    Article  PubMed Central  PubMed  Google Scholar 

  15. Tarin D. Clinical and biological implications of the tumor microenvironment. Cancer Microenviron. 2012;5(2):95–112.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  16. Akhtar M, Haider A, Rashid S, Al-Nabet ADMH. Paget’s “seed and soil” theory of cancer metastasis: an idea whose time has come. Adv Anat Pathol. 2019 Jan;26(1):69–74. https://doi.org/10.1097/PAP.0000000000000219.

    Article  CAS  PubMed  Google Scholar 

  17. Ewing J, editor. Neoplastic diseases: a treatise on tumours metastasis. 3rd ed. Philadelphia: Saunders; 1928. p. 76–88.

  18. Hart IR, Fidler IJ. Role of organ selectivity in the determination of metastatic patterns of B16 melanoma. Cancer Res. 1980;40:2281–7.

    CAS  PubMed  Google Scholar 

  19. Fabisiewicz A, Rodzos MS, Zaczek AJ, Grzybowska EA. Circulating tumor cells in early and advanced breast cancer: Biology and prognostic value. Int J Mol Sci. 2020;21:1671.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  20. Blank A, Roberts DE, Dawson H, Zlobec I, Lugli A. Tumor heterogeneity in primary colorectal cancer and corresponding metastases. Does the apple fall far from the tree? Front Med. 2018;5(234):1–8.

    Google Scholar 

  21. Vanharanta S, Massagué J. Origins of metastatic traits. Cancer cell. 2013;24(4):410–21.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  22. Testa U, Pelosi E, Castelli G. Colorectal cancer: genetic abnormalities, tumor progression, tumor heterogeneity, clonal evolution and tumor-initiating cells. Med Sci. 2018;6(2):31.

    Google Scholar 

  23. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.

    Article  CAS  PubMed  Google Scholar 

  24. Tallmadge JE, Fidler IJ. The biology of cancer metastasis: historical perspective. Cancer Res. 2010;70(14):5649–69.

    Article  Google Scholar 

  25. Pienta EJ, Robertson BA, Coffey DS, Taichman RS. The cancer diaspora: metastasis beyond the seed and soil hypothesis. Clin Cancer Res. 2013;19(21):5849–55.

    Article  PubMed  Google Scholar 

  26. Qian JJ, Akçay E. Competition and niche construction in a model of cancer metastasis. PLoS ONE. 2018;13(5):e0198163.

    Article  PubMed Central  PubMed  Google Scholar 

  27. Chiang AC, Massagué J. Molecular basis of metastasis. N Engl J Med. 2008;359(26):2814–23.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  28. Ren X, Kang B, Zhang Z. Understanding tumor ecosystems by single-cell sequencing: promises and limitations. Genome Biol. 2018;19:211.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  29. Ray SK, Mukherjee S. Nutrigenomics and lifestyle facet- a modulatory molecular evidence in progression of breast and colon cancer with emerging importance. Curr Mol Med. 2022;22(4):336–48. https://doi.org/10.2174/1566524021666210331151323.

    Article  CAS  PubMed  Google Scholar 

  30. Psaila B, Lyden D. The metastatic niche: adapting the foreign soil. Nat Rev Cancer. 2009;9:285–93.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  31. Birkbak NJ, McGranahan N. Cancer genome evolutionary trajectories in metastasis. Cancer cell. 2020;1(37):8–19.

    Article  Google Scholar 

  32. Allgayer H, Leupold JH, Patil N. Defining the “metastasome”: perspectives from the genome and molecular landscape in colorectal cancer for metastasis evolution and clinical consequences. Seminars Cancer Biol. 2020;60:1–13.

    Article  CAS  Google Scholar 

  33. Klein A. Parallel progression of primary tumours and metastases. Nature rev. 2009;9(4):302–12.

    CAS  Google Scholar 

  34. Bray LJ, Hutmacher DW, Bock N. Addressing patient specificity in the engineering of tumor models. Front BioengBiotechnol. 2019;7:217.

    Google Scholar 

  35. Mantovani F, Collavin L, Del Sal G. Mutant p53 as a guardian of the cancer cell. Cell Death Differ. 2019;26:199–212.

    Article  PubMed  Google Scholar 

  36. Ishaque N, Abba ML, Hauser C, Patil N, Paramasivam N, Huebschmann D, et al. Whole genome sequencing puts forward hypotheses on metastasis evolution and therapy in colorectal cancer. Nat Commun. 2018;9(1):4782.

    Article  ADS  PubMed Central  PubMed  Google Scholar 

  37. TachtsidisL MIM, Jacobsen N, Thompson EW, Saunders CM. Minimal residual disease in breast cancer: an overview of circulating and disseminated tumour cells. ClinExp Metastasis. 2016;33:521–50.

    Article  Google Scholar 

  38. Eyles J, Yang H, Abastado JP. Tumor cells disseminate early, but immune surveillance limits metastatic outgrowth, in a mouse model of melanoma. J Clin Invest. 2010;120(6):2030–9.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  39. Norton L, Massague J. Is cancer a disease of self-seeding? Nat Med. 2006;12(8):875–8.

    Article  CAS  PubMed  Google Scholar 

  40. Machnik M, Oleksiewicz U. Dynamic signatures of the epigenome: friend or foe? Cell. 2020;9:653. https://doi.org/10.3390/cells9030653.

    Article  CAS  Google Scholar 

  41. Welch DR, Hurst DR. Defining the hallmarks of metastasis. Cancer Res. 2019;79(12):3011–27.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  42. Leung ML, Davis A, Gao R, Casasent A, Wang Y, Sei E, et al. Single-cell DNA sequencing reveals a late-dissemination model in metastatic colorectal cancer. Genome Res. 2017;27(8):1287–99.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  43. Vermaat JS, Nijman IJ, Koudijs MJ, Gerritse FL, Scherer SJ, Mokry M, et al. Primary colorectal cancers and their subsequent hepatic metastases are genetically different: implications for selection of patients for targeted treatment. Clin Cancer Res. 2012;18(3):688–99.

    Article  CAS  PubMed  Google Scholar 

  44. Nicolazzo C, Raimondi C, Gradilone A, Emiliani A, Zeuner A, Francescangeli F. Circulating tumor cells in right- and left-sided colorectal cancer. Cancers. 2019;11(8):1042.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  45. Weigelt B, Peterse JL, Van’t veer LJ. Breast cancer metastasis: markers and models. Nat Rev Cancer. 2005;5(8):591–602.

    Article  CAS  PubMed  Google Scholar 

  46. Pantel K, Brakenhoff RH. Dissecting the metastatic cascade. Nat Rev Cancer. 2004;4(6):448–56.

    Article  CAS  PubMed  Google Scholar 

  47. Mehlen P, Puisieux A. Metastasis: a question of life or death. Nat Rev Cancer. 2006;6(6):449–58.

    Article  CAS  PubMed  Google Scholar 

  48. Swann JB, Smyth MJ. Immune surveillance of tumors. J Clin Invest. 2007;117(5):1137–46.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  49. Jukić MK, Luetić AT, Skudar-Lukinović V, Andreis I. The antimetastatic effect of macrophages restored by indomethacin: concomitant tumor immunity model. CollAntropol. 2010;34(3):899–904.

    Google Scholar 

  50. Bomken S, Fiser K, Heidenreich O, Vormoor J. Understanding the cancer stem cell. Br J Cancer. 2010;103(4):439–45.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  51. Oskarsson T, Batlle E, Massague J. Metastatic stem cells: sources, niches, and vital pathways. Cell stem cell. 2014;14(3):306–21.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  52. Youssef A, Aboalola D, Han VKM. The roles of insulin-like growth factors in mesenchymal stem cell niche stem cells. International. 2017:9453108. https://doi.org/10.1155/2017/9453108.

  53. Kim JH, Park SY, Jun Y, Kim JY, Nam JS. Roles of Wnt target genes in the journey of cancer stem cells. Int J Molecular Sci. 2017;18(8)

  54. Herrero MJ, Gitton Y. The untold stories of the speech gene, the FOXP2 cancer gene. Genes Cancer. 2018; https://doi.org/10.18632/genesandcancer.169.

  55. Liu C, Tang DG. MicroRNA regulation of cancer stem cells. Cancer Res. 2011;71(18):5950–4.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  56. Wang Z, Li Y, Sarkar FH. Notch signaling proteins: legitimate targets for cancer therapy. Curr Protein Pept Sci. 2010;11(6):398–408.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  57. Wynn TA, Chawla A, Pollard JW. Macrophage biology in development, homeostasis and disease. Nature. 2013;496:445–55.

    Article  ADS  CAS  PubMed Central  PubMed  Google Scholar 

  58. Chinen T, et al. An essential role for the IL-2 receptor in Treg cell function. Nat. Immunol. 2016;17:1322–33.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  59. Schwartz M, Zhang Y, Rosenblatt JD. B cell regulation of the anti- tumor response and role in carcinogenesis. J Immunother Cancer. 2016;4:40.

    Article  PubMed Central  PubMed  Google Scholar 

  60. Olkhanud PB, et al. Tumor- evoked regulatory B cells promote breast cancer metastasis by converting resting CD4+ T cells to T- regulatory cells. Cancer Res. 2011;71:3505–15.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  61. Chen DS, Mellman I. Oncology meets immunology: the cancer–immunity cycle. Immunity. 2013;39:1–10.

    Article  PubMed  Google Scholar 

  62. Pierobon M, Ramos C, Wong S, Hodge KA, Aldrich J, Byron S. Enrichment of PI3K-AKT–mTOR pathway activation in hepatic metastases from breast cancer. Clin Cancer Res. 2017;23(16):4919–28.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  63. Summy JM, Gallick GE. Src family kinases in tumor progression and metastasis. Cancer metastasis Rev. 2003;22(4):337–58.

    Article  CAS  PubMed  Google Scholar 

  64. Tsubakihara Y, AristidisMoustakas A. Epithelial-mesenchymal transition and metastasis under the control of transforming growth factor β. Int J Mol Sci. 2018;19(11):3672.

    Article  PubMed Central  PubMed  Google Scholar 

  65. Lamouille S, Xu J, Derynck R. Molecular mechanisms of epithelial–mesenchymal transition. Nat Rev Mol Cell Biol. 2014;15(3):178–96.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  66. Mudduluru G, Abba M, Batliner J, Patil N, Scharp M, Lunavat TR, et al. A systematic approach to defining the microRNA landscape in metastasis. Cancer Res. 2015;75(15):3010–9.

    Article  CAS  PubMed  Google Scholar 

  67. Heerboth S, Housman G, Leary M, Longacre M, Byler S, Lapinska K, et al. EMT and tumor metastasis. ClinTransl Med. 2015;4:6.

    Google Scholar 

  68. Vogelstein B, Kinzler KW. The multistep nature of cancer. TIG. 1993;9(4):138–41.

    Article  CAS  PubMed  Google Scholar 

  69. Arvelo F, Sojo F, Cotte C. Biology of colorectal cancer. ecancer. 2015;9:520. https://doi.org/10.3332/ecancer.2015.520.

    Article  Google Scholar 

  70. Lowdon RF, Wang T. Epigenomic annotation of noncoding mutations identifies mutated pathways in primary liver cancer. Plos One. 2017; https://doi.org/10.1371/journal.pone.0174032.

  71. Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad M, et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med. 2018;24(5):541–50.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  72. Müller A, Homey B, Soto H, Ge N, Catron D, Buchanan ME, et al. Involvement of chemokine receptors in breast cancer metastasis. Nature. 2001;410(6824):50–6.

    Article  ADS  PubMed  Google Scholar 

  73. Zlotnik A. New insights on the role of CXCR4 in cancer metastasis. J Pathol. 2008;215(3):211–3.

    Article  CAS  PubMed  Google Scholar 

  74. Fleming M, Ravula S, Tatishchev SF, Wang HL. Colorectal carcinoma: pathologic aspects. J GastrointestOncol. 2012;3(3):153–73.

    Google Scholar 

  75. Condeelis J, Weissleder R. In vivo imaging in cancer cold spring. HarbPerspect Biol. 2010;2(12):a003848.

    CAS  Google Scholar 

  76. Menezes ME, Das SK, Minn I, Emdad L, Wang XY, Sarkar D, et al. Detecting tumor metastases: the road to therapy starts here. Adv Cancer Res. 2016;132:1–44.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  77. Foncillas JG, Sunakawa Y, Aderka D, Wainberg Z, Ronga P, Witzler P, et al. Distinguishing features of cetuximab and panitumumab in colorectal cancer and other solid tumors. Front Oncol. 2019;9:849.

    Article  Google Scholar 

  78. McCord M, Mukouyama Y, Gilbert MR, Jackson S. Targeting WNT signaling for multifaceted glioblastoma therapy Front. Cell. Neurosci. 2017; https://doi.org/10.3389/fncel.2017.00318.

  79. Li X, Wu Y, Tian T. TGF-β signaling in metastatic colorectal cancer (mCRC): from underlying mechanism to potential applications in clinical development. Int J Mol Sci. 2022 Nov 20;23(22):14436.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  80. Parseghian CM, Parikh NU, Wu JY, Jiang ZQ, Henderson L, Tian F, et al. Dual inhibition of EGFR and c-src by cetuximab and dasatinib combined with FOLFOX chemotherapy in patients with metastatic colorectal cancer. Clin Cancer Res. 2017;23(15):4146–54.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  81. Janssen LME, Ramsay EE, Logsdon CD, Overwijk WW. The immune system in cancer metastasis: friend or foe? J Immuno Therap Cancer. 2017;5:79.

    Article  Google Scholar 

  82. Celiá-Terrassa T, Kang Y. Metastatic niche functions and therapeutic opportunities. Nature Cell Biol. 2018;20(8):868–77.

    Article  PubMed  Google Scholar 

  83. Ray SK, Mukherjee S. Cell free DNA as an evolving liquid biopsy biomarker for initial diagnosis and therapeutic nursing in cancer- an evolving aspect in medical biotechnology. Curr Pharm Biotechnol. 2022;23(1):112–22. https://doi.org/10.2174/1389201021666201211102710.

    Article  CAS  PubMed  Google Scholar 

  84. Ray SK, Mukherjee S. Current headway in cancer immunotherapy emphasizing the practice of genetically engineered T cells to target selected tumor antigens. Crit Rev Immunol. 2021;41(1):23–40. https://doi.org/10.1615/CritRevImmunol.2020037044.

    Article  PubMed  Google Scholar 

  85. Ray SK, Mukherjee S. Cancer stem cells: current status and therapeutic implications in cancer therapy-a new paradigm. Curr Stem Cell Res Ther. 2021;16(8):970–9. https://doi.org/10.2174/1574888X16666210203105800.

    Article  CAS  PubMed  Google Scholar 

  86. Ray SK, Mukherjee S. Consequences of extracellular matrix remodeling in headway and metastasis of cancer along with novel immunotherapies: a great promise for future endeavor. Anticancer Agents Med Chem. 2022;22(7):1257–71. https://doi.org/10.2174/1871520621666210712090017.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Declared none

Funding

None

Author information

Authors and Affiliations

Authors

Contributions

SKR and SM conceptualized the manuscript. SKR wrote the first draft. SM edited it. SKR drew the figures. Both authors approved the final manuscript.

Corresponding author

Correspondence to Sukhes Mukherjee.

Ethics declarations

Ethics and consent to participate

Not applicable

Consent for publication

Not applicable

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ray, S.K., Mukherjee, S. Molecular perspectives on systemic priming and concomitant immunity in colorectal carcinoma. J Egypt Natl Canc Inst 36, 7 (2024). https://doi.org/10.1186/s43046-024-00211-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43046-024-00211-9

Keywords