Summary
Cancer is a disease that stems from a fundamental liability inherent to multicellular life forms in which an individual cell is capable of reneging on the interest of the collective organism. While cancer is commonly described as an evolutionary process, a less appreciated aspect of tumorigenesis may be the constraints imposed by the organism’s developmental programs. Recent work from single-cell transcriptomic analyses across 24 cancer types has revealed the recurrence, plasticity, and co-option of distinct cellular states among cancer cell populations. Here, we note that across diverse cancer types, the observed cell states are proximate within the developmental hierarchy of the cell-of-origin. We thus posit a model by which cancer cell states are directly constrained by the organisms’ ‘developmental map’. According to this model, as a population of cancer cells traverses the developmental map, it generates a heterogeneous set of states whose interactions underpin emergent tumor behavior.
In Brief
Single-cell transcriptomic analyses in diverse cancer types have revealed the recurrence, plasticity and co-option of distinct cellular states, leading to a “developmental constraint” model whereby cancer cell state plasticity is restricted by the organism’s developmental map to access progenitor-like states and differentiated-like states of adjacent lineages.
The tumor is a highly complex organ composed of myriad cell types, each in a range of cellular states1. The advent of single-cell RNA-seq has permitted a global view of the transcriptomes of cancer and microenvironment cells across individual tumors2,3. Such transcriptomic analyses have enabled the identification of resident cell types as discontinuous clusters, as well as a continuum within an individual cluster indicating diverse cellular states4,5. Over the past decade, transcriptional variation has been identified within the malignant cell compartment across 24 cancer types in over 80 different studies6–8. Importantly, this variation within the malignant cell population comes in the form of functionally-related sets of differentially expressed genes. For example, in cancer cells of lung adenocarcinoma tumors, one set of differentially expressed genes – a gene module – includes CAV1, AGER and LMO7; genes which were previously described as important markers of the alveolar type I differentiation cell state8. Cancer cells expressing this alveolar differentiation gene module were found to be associated with better survival in lung cancer patients with residual disease9. This relationship between the expression of gene modules in cancer cells and the fate of the patient underscores the importance of understanding their emergence and role in the tumor.
Notably, an individual cell is not necessarily restricted to the expression of a single gene module but rather several modules may be expressed in conjunction to achieve a range of phenotypic properties2. Thus, a cancer cell’s state appears to be assembled by the heterogeneous expression of gene modules8,10. From single cell transcriptomic analyses of an individual patient’s tumor we generally find, among the cancer cells, overwhelming evidence for a range of states2, pointing to transcriptional heterogeneity as a critical characteristic of tumor biology.
In this Perspective, we provide a conceptual framework to synthesize the findings from published single-cell transcriptomic atlases and propose a unified model on the causes and constraints of tumor heterogeneity. This view provides a pathway from the descriptive characterization of cancer cell states across distinct cancer types towards a more mechanistic understanding of the developmental pressures acting on a tumor’s composition of cell states. Interrogating the developmental constraints inherent to a tumor’s cellular states offers a research program poised to lead to new therapeutic strategies and improved clinical outcomes.
Cell state recurrence, co-option of programs and cellular plasticity
Extensive work on the transcriptional variation among cancer cells has revealed three important properties. First, the gene modules underpinning states recur across tumors of the same cancer type11, and even across cancer types8. In lung adenocarcinoma, tumors with distinct driver mutations (EGFR, KRAS and MET) transcriptionally converge to a population of cells in the alveolar, ciliated-like and proliferative states12. Similarly, across the three genetic subtypes of breast cancer (ER+, HER2+ and triple-negative) malignant cells share common cancer cell states13,14. Moreover, in glioblastoma10, prostate cancer15,16, ovarian17 and colorectal cancer18, analyses of tumors with distinct driver mutations have revealed a convergence of transcriptional heterogeneity, indicating that the repertoire of cancer cell states is a fundamental property of tumorigenesis. This, in turn, implies conservation of a range of functional properties that may be required to sustain tumors and fuel progression.
Second, the recurrent gene modules are mostly not uniquely expressed in cancer, but rather shared with gene modules induced in normal physiology and tissue homeostasis. One class of these shared – or co-opted – gene modules relate to biological processes such as stress19, the interferon response20, and hypoxia programs21. The stress-like cell state occurs at a higher frequency among cancer cell populations than non-cancer ones19. Pan-cancer studies defined a catalog of recurrent gene modules, including one with the genes ISG15, ISG20, IFI6, HLA-DR; which are collectively expressed in the interferon response7,8. It has also been shown that cancer cell-derived interferons regulate functional polarization of tumor-associated monocytes, predicting a better response to immunotherapy20.
Beyond these biological process-related gene modules, a second class of gene modules corresponds to cell identity. For example, the pigmented cell state in melanoma expresses melanocytic gene programs of the skin22, and mature luminal-like cell states in breast cancer express programs of luminal cells in the breast13. Collectively then, the gene modules underlying the cancer cell states appear to be co-opted from developmental and differentiation programs. The cancer cell-of-origin is co-opted in many cancers, in addition to the co-option of states reminiscent of other tissue specific cell types, as we discuss below.
A third crucial aspect of cancer cell states is their potential to transition between states by invoking cellular plasticity. Using single-cell and lineage tracing technologies, studies in a number of cancer types have revealed the occurrence of remarkable cellular plasticity during tumor evolution23. Murine lung adenocarcinoma comprises a transcriptionally distinct subpopulation of ‘highly plastic cancer cells’, which when transplanted into a syngeneic mouse are able to generate a tumor that reconstitutes the full transcriptional heterogeneity of the original tumor24. In vivo studies have also revealed the switching between the invasive and proliferative subtypes of melanoma, supporting plasticity between melanoma cell states25. In rhabdomyosarcomas, the mesenchymal cancer cell state has been described as having tumor initiating capacity and inherent plasticity to recapitulate all the states of the initial rhabdomyosarcoma tumor26. In glioblastoma, Neftel et al. isolated two of the four dominant glioblastoma states – mesenchymal-like and neural-progenitor like – and implanted subpopulations of three of the four dominant glioblastoma states – astrocyte-like, mesenchymal-like, and neural-progenitor like – revealing that these individual cell identity cancer states have tumor-initiating capacity and lead to tumors with a reconstitution of all four neuronal states10. Furthermore, using an in vivo barcoding experiment the authors suggest that all four glioblastoma cell states have the capacity to differentiate to other states.
The ability for individual cancer cells to reconstitute a tumor with the transcriptional diversity of the original demonstrates the highly plastic nature of cancer cell states, as well as their general non-genetic dependencies. In the same glioblastoma study10, tumors with distinct genetic alterations (EGFR, CDK4, NF1, and PDGFRA) were generally composed of the same four neuronal cell identity cancer cell states, though at different frequencies. For example, in tumors with EGFR amplifications, the frequency of cells in the astrocyte-like cell state was higher than that in tumors with other genetic alterations. Furthermore, genetic alterations in basal cells or luminal cells induce cell state changes, as has been shown in basal cell carcinoma27,28. The effect on cancer cell state frequencies as a function of the genetic alterations suggests an interaction between these two basic mechanisms. We may refer to such a process as ‘plastigenic’, as it involves both genetic and non-genetic contributions. This is observed in gliomas, where IDH-mutant and IDH-wildtype gliomas harbor differential capacities for cellular plasticity29. Interestingly, a plastigenic process reliant upon both genetic changes and the cell’s basic abilities for plasticity may be required for tumorigenesis.
Correspondence between cancer cell states and the developmental map
The recurrence, co-option and plasticity of cancer cell states has led us to note an apparently general pattern relating to their occurrence: within a given cancer type, the observed cell states are related along the developmental hierarchy. Consider melanoma, where it has been long known that gene expression programs can distinguish particular subtypes30,31. Melanoma tumors have been classified into either the invasive/undifferentiated subtype, marked by the expression of AXL32, or the differentiated/proliferative subtype marked by the expression of melanocyte lineage factor MITF33. Beyond the expression of marker genes, single-cell analyses have shown that melanoma cancer cells express cell type specific gene programs and are present in entirely distinct cell identity cellular states. Among these states are (1) a differentiated melanocyte cell state, (2) a neural crest-like cell state, and (3) an invasive/mesenchymal-like cell state22,34–36. Importantly, the normal counterparts of these cell identity states are directly adjacent in the developmental map, according to which the cell types originate. Melanoblasts – thought to be the cell-of-origin for melanoma – originate developmentally from the ectoderm37, which gives rise to neural crest cells38. The states that recur in melanoma cancer cells thus correspond precisely to the developmentally adjacent cell states of the melanoblasts cell-type of origin.
Lung adenocarcinoma tumors originating from alveolar type II cells exhibit 7 cancer cell states12,24,39,40: the (1) alveolar type I-like cell state enriched with the expression of alveolar type-I specific genes such as Ager, Cav1, and Hopx, (2) the alveolar type II-like marked by the expression of surfactant proteins Sftpc, Sftpb and Napsa, among others, (3) the mixed-alveolar-type I/II cell state expressing mixed lineage genes such as Muc5b, Nkx2–1, and Pdpn, (4) a club-like cell state that expresses secretary genes such as Scgb3a1 and Scgb3a2, (5) an embryonic-liver-like cell state marked by the expression of genes including Cldn2, and Dlk1, (6) a gastric- or gut-like cell state expressing genes such as Dgat2 and Itih2, and (7) an EMT cell state expressing the canonical markers such as Vim and Zeb1. Applications of single-cell transcriptomic and multiplex immunofluorescence have identified cell identity states along the developmental trajectory specifically in primary human lung adenocarcinoma tumors12,39 as well as in pan-cancer studies7,8. Five of these states are marked by the expression of gene expression programs from cell types within the lung tissue: the alveolar type I-like, alveolar type II-like, mixed-alveolar-type I/II, club and ciliated cell states, that map developmentally to cells derived from a common lung progenitor41,42 (Figure 1). The gastric-like or gut cell-like state expressed gene modules of a developmentally primitive cell state – the foregut progenitor cell state. Finally, the embryonic-liver gene program24,43 is enriched for the expression of a more distant cell-type that is developmentally accessible through the primitive gut progenitor state. These observations suggest that malignant cells in lung adenocarcinoma navigate both in the ‘reverse’ direction to de-differentiate towards progenitor cell states and ‘forward’ direction along the developmental map to differentiate to mature cell states.
Figure 1. Correspondence between lung cell types in development and cancer cell states in lung adenocarcinoma.
a, An illustration of dimensionality reduction projections of single-cell transcriptomic data from normal lung112 and mouse embryonic developmental atlases113, which were used to generate the developmental hierarchy of cell types shown in c. b, Same as a, for cell identity-related cancer cell states in lung adenocarcinoma, as curated from single-cell transcriptomic studies12,24,39. c, (Top) A simplified developmental hierarchy of cell types identified using the approach diagrammed in a, within the foregut branch. Marker genes are indicated for relevant developmental cell types. (Bottom) Lung adenocarcinoma cancer cell states identified using the approach diagrammed in b, are arranged according to the developmental hierarchy, matched by corresponding gene expression programs.
Recent work from Desai and colleagues has revealed that transgene activation in alveolar type I cells also leads to adenomatous transformation with a more indolent histology44. At early stages of tumorigenesis, the tumors originating from alveolar type I cells transition through an intermediate state expressing both alveolar type I and alveolar type II marker genes that are normally mutually exclusive44. During later stages of tumor development, the tumors harbor states expressing lung progenitor cell states and gastric cell states, reminiscent of cell states derived from alveolar type II cells44. Thus, irrespective of the cell-of-origin, lung adenocarcinoma cancer cells appear able to traverse the foregut developmental branch to assume distinct states through developmentally restricted cellular plasticity.
In tumors of lung squamous cell carcinoma – another histological subtype of lung cancer – cancer cell states recapitulate the cell identities derived from the proximal lung progenitor, including the basal-like cell state, the ciliated-like cell state and the club-like state45. Notably both lung histology tumors share the club-like and ciliated-like cell state (Figure 2).
Figure 2. The developmental map constrains the occurrence of cancer cell states in seven cancer types.
(Top) A simplified developmental lineage of cell types based on a singlecell transcriptomic atlas of mouse embryonic development45 and tissue-specific reviews for pancreas,48 lung,41,42 skin,37,38 brain,49 muscles, and blood lineages.50 (Bottom) A synthesis of cancer cell states as detected in 7 cancer types. To generate this map, we first curated a list of single-cell transcriptomic atlases for each cancer type: pancreatic ductal adenocarcinoma,51,52 lung adenocarcinoma,12,24,39 lung squamous cell carcinoma,47 melanoma,22,34,35,36 glioblastoma,10 acute myeloid leukemia,53,54,55 and rhabdomyosarcomas.26 Second, we curated lists of the identified cancer cell states that are related to cell identity and excluded those related to biological processes (such as cycling and stress). Finally, we arranged the cancer cell states in accordance with the developmental map. Arrows indicate evidenced cell state transitions. Stars indicate the purported cell of origin. Developmental cell type and cancer cell states are colored by cancer type.
In glioblastoma tumors, Neftel et al. provided evidence that cancer cells assumed either an astrocyte progenitor-like state (APC), a neural progenitor-like state (NPC), an oligodendrocyte progenitor-like state (OPC) or a mesenchymal like cancer cell state (MES)10. Other studies have revealed a more granular classification of these cell states46,47. The APC-like and OPC-like states are transcriptomically similar to the astrocyte progenitor cell and oligodendrocyte progenitor cell, respectively, and these are developmentally adjacent, deriving from a common glial progenitor cell48. The NPC state is marked by gene expression programs similar to the neural progenitor cell that is developmentally upstream on the developmental map48 (Figure 2). While in glioblastoma the cell-of-origin remains to be one of the major unresolved questions49,50, the putative cell types are the oligodendrocyte progenitor cell47,51 and differentiated neurons52. Thus, the glioblastoma cell state plasticity programs apparently enable malignant cells to not only assume upstream stem-like states, but also those differentiated states accessible through multiple common upstream progenitor states. If we assume that the oligodendrocyte progenitor cell is the cell-of-origin, the observed cell state heterogeneity suggests that the malignant OPC-like cells are constrained to traverse the map to exploit accessible states both in ‘reverse’ mode to de-differentiate into the NPC-like state as well as in ‘forward’ mode to differentiate into the APC-like state. Alternatively, if a differentiated neuron is the cell-of-origin for a glioblastoma, the malignant cell states would still traverse the developmental map in the ‘reverse’ mode to de-differentiate into the NPC-like state and the ‘forward’ mode to differentiate into the APC-like and OPC-like state.
Pancreatic ductal adenocarcinoma has been historically defined transcriptionally as either ‘classical’ – tumors with pancreatic progenitor gene expression programs – or ‘basal-like’ – tumors with basaloid or mesenchymal-like transcriptional programs53–55. Single-cell studies have added insight into this classification by revealing that individual tumors simultaneously harbor subpopulations of both states56. Specifically, these studies have identified (1) a cancer cell state co-opting the acinar cell program of the pancreas (acinar-like), (2) a classical cancer cell state expressing gene programs of the pancreatic progenitor cell and (3) a neuroendocrine-progenitor like cell state that shares transcriptional similarities to the endocrine progenitor in the pancreas57,58 (Figure 2). The authors also identified two additional intriguing cell states: the basaloid and the squamoid57 (Figure 2). Recent work has elucidated that large pancreatic ducts comprise dNp63 expressing basal cell-types59, however, the developmental trajectory of these cells and the trajectory that tumor cells take to acquire these states are still being explored60. Examining the developmental map, the basal and squamous cells of the esophagus are derived within the same foregut developmental path as the pancreas61. One prediction is thus that pancreatic cells assume the basal and squamous states by traversing the map through early pancreatic-esophageal progenitor states.
In mesoderm-originating tumors, a similar correspondence is found between the cell states of the tumor and those within the developmental lineage. For example, in rhabdomyosarcomas, the cell-of-origin is striated muscle and its tumors harbor (1) a subpopulation of cells in the differentiated muscle cell state (expressing genes such as like MYLPF, ACTC1, and TSPAN3), (2) a mesenchymal-enriched population (expressing extracellular matrix and mesenchymal genes including MMP2, CD44, PTN, POSTN and THY1), and (3) ‘ground state’-like cells reflecting a similarity to a muscle progenitor cell, which express muscle-lineage specific genes such as MYOD, MYOG and Desmin (indicating a commitment to the muscle lineage), but not specifically inducing myosin26. Again, it is evident that the corresponding developmental stages are adjacent along the developmental map (Figure 2).
Landmark studies in acute myeloid leukemia (AML) have demonstrated the heterogeneity in this cancer type by identifying populations of cells with distinct cell surface marker expression62. Two main subpopulations have been recognized: (1) primitive AML cells or leukemic stem cells that sustain the tumor and promote therapeutic resistance and (2) differentiated AML cells that may retain hematological functions to create pathological interactions with the tumor microenvironment63. While it was initially believed that transcription factor alterations in AML led to ‘differentiation blocks’64, whereby progenitor states are locked-in, the application of single-cell RNA-seq has now revealed that malignant AML cells transcriptionally mimic features of cell states along the myeloid development hierarchy65. During normal hematopoiesis, hematopoietic stem cells give rise to lineage-committed progenitor states, including the common lymphoid progenitor and common myeloid progenitor that ultimately give rise to mature cell types66. AML tumors encompass cell states that recapitulate both progenitor states such as the hematopoietic stem-like state, the progenitor-like state and differentiated cell types such as the cDC-like cell state and the monocyte-like cell state65,67,68. Consistently, cell states in AML tumors map to the developmental stages of hematopoiesis.
Notably, the ‘mesenchymal-like’ cell state appears to recur across different cancer types69. Epithelial malignant cells often deploy a transition to a mesenchymal-like state to acquire motility and invasive properties by invoking the developmental program for an epithelial-to-mesenchymal transition (EMT)70. While the mesenchymal-state recurs across cancer types, a recent pan-cancer study described five non-overlapping gene expression programs that independently describe a mesenchymal state. These EMT or mesenchymal-like gene expression programs vary with specificity to distinct cancer types7. This suggests that while there may be shared features of the mesenchymal state across tumor types, such as the expression of canonical marker Vimentin71, it is possible the cells retain a memory of its developmental history. Supporting the notion that the mesenchymal states encode tissue specific memory, are observations from reconstitution experiments, where the mesenchymal state from rhabdomyosarcoma and glioblastoma tumors is isolated and injected into a mouse the resulting tumors recapitulate the histology and cell state diversity of the original tumor, indicating the retention of elements from the original cancer initiating cell type10,26. Furthermore, comparative analyses of the transcriptional dynamics during EMT have revealed that the transition to the mesenchymal cell state is largely context-dependent69, and indeed the context-specificity of ‘EMT genes’ was also observed in primary tumors72. Another study developed a mesenchymal-stromal decoupling approach that revealed three distinct patterns of mesenchymal gene expression programs in squamous, gynecological and gastrointestinal tumors73. Collectively, it appears that the mesenchymal state may be best interpreted as a common cancer motif71 with context-specific features involving contributions from developmental, epigenetic and genetic factors.
The developmental constraint model
As discussed above, support for the notion that cancer cell states co-opt developmental states comes from studies across several cancer types that have identified similar cell identity related states through independent analytical methods and datasets. Based upon the composition of the malignant cellular states in the seven cancer types described in Figure 2, it seems evident that the appearance of cancer cell states is constrained by the underlying developmental map. We thus propose a ‘developmental constraint’ model, whereby the malignant population – originating from the initial state of the cell-of-origin – traverses the states accessible by their developmental trajectory, both toward progenitor-like states and differentiated-like states of adjacent lineages, collectively forming a heterogeneous population. Specifically, the developmental constraint model restricts cell states transitions to occur only among states directly related in the developmental map.
The developmental constraint model indicates the importance of the cancer cell-of-origin in determining the cell state content of the heterogeneous malignant population. While for a number of cancer types the cell-of-origin remains unknown74, in a few cancer types the cell-of-origin is known from studies of genetically engineered mouse models with oncogene activation. In lung adenocarcinoma, for example, where Kras activation in alveolar type II cells initiates tumors75, the observed states are consistent with the notion of a cancer population traversing the developmental map to spawn the related cellular states (Figure 1).
Further support for the model derives from the evidence that tumors in a given tissue give rise to distinct cell states depending upon the precise cell-of-origin. Lung adenocarcinomas and lung squamous cell carcinoma tumors, for example, originate from developmentally distant cell types – alveolar type I/II and basal cells, respectively. Consistently, lung squamous cell carcinoma cell tumors co-opt the states from the proximal lung cell types while lung adenocarcinoma co-opt the states lung alveolar cell types. We note that in mouse models of lung squamous cell carcinoma Sox2 activation in combination with loss of Cdkn2ab and Pten in both alveolar type II or basal cells generate tumors with lung squamous cell carcinoma histology, however their specific patterns of cell state heterogeneity remain unknown76. Therefore, notwithstanding the potency of driver mutations to determine tumor histology, the specific pattern of cell state heterogeneity may be determined by the tumor’s cell-of-origin.
Molecular encodings of developmental constraints
The developmental constraint model may enable insight into the function of the co-opted states in tumorigenesis, by studying the underlying molecular mechanisms and by reference to their roles within developmental pathways. The developmental constraints that restrict cell state transitions along the developmental map likely include aspects of chromatin remodeling required for cell state transitions. This follows from the observation that lineage-commitment programs in embryonic development and differentiation are typically accompanied by highly dynamic chromatin changes77. For example, in the hematopoietic system, a population of hematopoietic stem cells gives rise to a variety of cell types by a specific sets of transcription factors and chromatin remodelers that establish regulatory chromatin states and subsequently lead to lineage differentiated cells78,79. Analogously, during tumorigenesis, as an oncogene-transformed cancer cell-of-origin establishes a tumor, it is likely that chromatin changes enable the generation of tumor heterogeneity80. We speculate that cancer cells rewire their chromatin state to a plasticity-permissive state that is epigenetically reminiscent of a developmental cell type.
Chromatin remodelers such as histone-lysine demethylases are frequently mutated in cancer and can promote activation of gene expression programs allowing bidirectional cell state transitions81. Furthermore, gain-of-function EZH2 mutations in lymphomas and myelomas alter the regulatory genome by expansive Histone3-Lys27 trimethylation that restrict the cells from specific cell states82,83. More recently, Loukas et al. found that disrupting the epigenetic regulatory network led to a competitive advantage for cancer cells in different tumor microenvironments84. Additionally, a number of studies have revealed the role of chromatin modifiers in establishing and regulating cancer cell states. For example, the histone demethylase PHF8 establishes a metastatic and invasive melanoma cell state85 and inhibition of KDM1A, another histone demethylase, suppresses a neuroendocrine cell state in small cell lung cancer86. We thus speculate that transcription factors and chromatin remodelers enable co-option of developmental epigenetic networks to establish specific regulatory elements such as bivalent promoters87,88 and chromatin loops to reprogram enhancer-promoter contacts89. Consequently, identifying chromatin accessibility-imposed constraints on a cell’s identity may reveal the principles by which the developmental map is encoded and exploited by the malignant cell population.
Relationship between the developmental constraint model and existing models
One long-discussed notion describes cancer as a de-differentiation process, suggesting ‘development in reverse’70. While a transition to stem-like states is common between this and the developmental constraint model, the de-differentiation model does not account for the possibility of also assuming more differentiated states that are distinct from the cell-of-origin. As we have noted, in glioblastomas and lung adenocarcinoma the cancer cell population invokes several states within the accessible developmental map and acquires states that require ‘forward differentiation’.
The cancer stem cell (CSC) concept is another prominent model for tumor growth90,91. Under this model, tumors are composed of sub-populations that are related by a hierarchy90. At its apex, is a stem cell or stem cell-like population, whose proliferation leads to cellular states that ‘roll down’ to assume differentiated states92. A crucial aspect of the CSC model is that the differentiated states have a limited propagation potential, such that only the CSCs are able to initiate new tumors. Indeed, Tirosh et al. in their study of oligodendroglioma identified a stem-like state and described it as evidence for the CSC model11.
Both the CSC and developmental constraint models agree on the occurrence of cell state heterogeneity and its maintenance. According to the CSC model, cellular heterogeneity results from stem-like cancer cells differentiating into non-CSC malignant cells. In contrast, in the developmental constraint model, heterogeneity occurs as the malignant cell population traverses in both directions: forward towards differentiation and in reverse, de-differentiating towards stem-like states. This is supported by studies in colorectal cancer that revealed the Lgr5+ stem cell population is replenished by Lgr5-cancer cells93 and in melanoma, revealing that cancer cell states transition between identities to enable metastasis and tumor growth35. Conceptually, it suggests that cancer cell states – rather than being organized hierarchically – can each access their inherent plasticity to traverse the available states within the constraints of the developmental map.
As we have reviewed above, recent work has revealed that the bulk of the tumor consists of rapidly proliferating and differentiated cell states94. Moreover, there is evidence that distinct states – including stem-like states – are able to reconstitute the tumor by cellular plasticity. Essentially then, the developmental constraint model provides a contrasting model centered around ‘plasticity’ by which no population holds a privileged propagating potential, and operating within the contexts of developmental constraints that act to limit the range of possible cellular states.
Another related concept is lineage infidelity – or trans-differentiation – whereby a tumor converts from one histological subtype to another. Trans-differentiation has been most notably observed in lung and prostate cancer that enables resistance to therapy95. Histological trans-differentiation of lung adenocarcinomas to aggressive neuroendocrine derivative (small-cell lung cancer) frequently occurs in patients with EGFR mutant lung adenocarcinoma treated with targeted therapy against mutant EGFR96,97. Most recently, clinical and retrospective studies have reported that lung adenocarcinoma patients treated with targeted therapy against EGFR and KRAS mutations have demonstrated squamous transformation98,99. During the lung adenocarcinoma to lung squamous cell carcinoma histological transition, cancer cell heterogeneity transitions from cells in ‘alveolar’ cell states to cancer cells expressing basal-cell related gene modules through a multi-lineage cell state100. This can be readily explained by the developmental constraint model, as alveolar cell states are separated from basal cell states by multiple upstream progenitor states, though still within the same lung progenitor developmental branch (Figure 2). Similarly, in prostate cancer, under therapeutic pressure, such as anti-androgen therapy, prostate adenocarcinomas trans-differentiate to neuroendocrine cancers101; acquiring states within the same developmental lineages102.
Our model posits a strong constraint on the establishment of new states, even under intense evolutionary pressures such as therapeutic intervention. The mechanisms underlying trans-differentiation are an active area of research, and it will be interesting to study how therapeutic intervention remains constrained by the developmental lineage.
Predictions and limitations of the developmental constraint model
How the heterogeneity in the malignant population confers functional relationships at the tumor-system level still remains elusive. The developmental constraint model predicts that the cancer cell states may be co-opted for large-scale spatial organization to achieve the physiological requirements of the tumor. From developmental biology it is known that setting up the embryonic axes and cell type specification in epiblast derivatives during gastrulation occurs by signaling molecule gradients and transcription factor activity. A number of studies have shown that cells are allocated to specific fates according to their spatio-temporal position in the streak103. Specifically, the nodal–SMAD2 and SMAD3 signaling pathway104 and the canonical Wnt signaling pathway105 are critical in the posterior-distal axis. We also have a detailed understanding of the mechanisms by which the “toolkit genes” are deployed to set up morphogenic signals (such as bicoid in Drosophila) and the gene circuits leading to the patterning106. Cancer cells may co-opt such developmental pathways to set up and maintain spatial axes of orientation among the states accessible from the developmental map. This co-option has been observed in teratomas, malignant transformation of germ cells, where organogenesis and spatial patterning allows for the formation of entire organs such as teeth, bone and other structures107. Morphogenic signals may also be co-opted by the cancer population to particular individual states or maintain the heterogeneity. Such collective behavior may function to allow the malignant population to set up evasion programs, whereby an axis through the tumor may allow cells facing the immune system to adopt immune-evasive cell states while tumor cells within the core adopt a more proliferative state8,108. Spatial transcriptomics studies are uncovering structural relationships among the tumor’s cancer cell states, indicating plausibility of higher-order architectures109. Additional overlapping codes may specify axes of stem-like states, migration, and interactions with elements of the tumor microenvironment such as fibroblasts110.
The relationship between cancer cell states and the developmental map may shed light on mechanisms of drug tolerance with direct impact on patient outcomes. A central aspect to the developmental constraint model is the transitions between cancer cell states. One application of this aspect as a new therapeutic direction would thus be to inhibit developmentally constrained cell state transitions. Inhibiting cellular plasticity would seek to reduce the heterogeneity of cell states present in the tumor compartment. Indeed, efforts to inhibit cell state transitions have revealed that inhibiting HDAC1 can suppress the acinar-to-ductal plasticity in pancreatic cancer, providing a therapeutic opportunity for pancreatic ductal adenocarcinoma patients111. Furthermore, inhibiting developmentally constrained cell state transitions may provide an opportunity to restrict adaptation to drug treatment and mechanisms of acquired resistance.
Another prediction of the developmental constraint model would be to better assess the stage of the tumor. Progression along the map appears to be related to the aggressiveness of the tumor, as, for example, neuroblastoma is an undifferentiated and highly aggressive tumor, whereas ganglioneuroma is differentiated and essentially a benign version. Through a better understanding of the obstacles and assistance that cancer cells face we may be able to exploit a tumor’s cell state composition to direct and refine therapeutics for improved patient outcomes.
One limitation of the developmental constraint model is that it does not describe the extent to which the cancer cell states may travel along the developmental map. For example, the model does not explain the restraints on AML cancer cells that restrict differentiation to the states along the lineage’s states. It may thus be that there are additional lineage constraints at the chromatin level or cancer-specific features that prevent the accessing by the cancer of developmental states that are in principle available to it. It remains unknown, however, whether the malignant cells of a tumor cannot access such developmental states or whether these simply do not provide the cancer cell with a competitive advantage in the specific tumor environment.
Overall, the developmental constraint model provides insight into the unifying constraints and strategies that establish the composition of the malignant cell states across tumor types. Among its strengths are its ability to make testable predictions about the set of states that compose a tumor with a particular cell of origin, as well as a tumor’s stage of progression given only its state composition. This view of the constraints imposed on tumor heterogeneity is supported by observations regarding cellular states, however, further work is required to incorporate the mechanistic dependencies of the cell state transitions, their relationship to the tumor spatial architecture, and interactions with the microenvironment. It will be particularly interesting to reveal the adaptive pressures that maintain the heterogeneity of the distinct functional states and their role in tumor physiology.
Highlights.
Not relevant for the Perspective format.
Acknowledgements
We thank Rich White, Amanda Lund, Sergei Doulatov, Gustavo França, Debbie Liberman, Dalia Barkley, Maayan Pour, Andrew Pountain, Tuomas Tammela, Yitzhak Pilpel and members of the Yanai lab for their helpful comments. We also thank the three anonymous reviewers for important comments. This work was supported by NIH grants U01CA260432, R01LM013522, R21CA264361 and U54CA263001 (I.Y).
Footnotes
Declaration of Interests
The authors declare no conflicts of interests.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Binnewies M. et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat. Med 24, 541–550 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Barkley D, Rao A, Pour M, França GS & Yanai I. Cancer cell states and emergent properties of the dynamic tumor system. Genome Res. 31, 1719–1727 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Potter SS Single-cell RNA sequencing for the study of development, physiology and disease. Nat. Rev. Nephrol 14, 479–492 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Xia B. & Yanai I. A periodic table of cell types. Development 146, (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Heumos L. et al. Best practices for single-cell analysis across modalities. Nat. Rev. Genet 24, 550–572 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Patel AP et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science 344, 1396–1401 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Gavish A. et al. Hallmarks of transcriptional intratumour heterogeneity across a thousand tumours. Nature 618, 598–606 (2023). [DOI] [PubMed] [Google Scholar]
- 8.Barkley D. et al. Cancer cell states recur across tumor types and form specific interactions with the tumor microenvironment. Nat. Genet 54, 1192–1201 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Maynard A. et al. Therapy-Induced Evolution of Human Lung Cancer Revealed by Single-Cell RNA Sequencing. Cell 182, 1232–1251.e22 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Neftel C, et al. An Integrative Model of Cellular States, Plasticity, and Genetics for Glioblastoma. Cell 178, 835–849.e21. (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Tirosh I. et al. Single-cell RNA-seq supports a developmental hierarchy in human oligodendroglioma. Nature 539, 309–313 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Kim N. et al. Single-cell RNA sequencing demonstrates the molecular and cellular reprogramming of metastatic lung adenocarcinoma. Nat. Commun 11, 1–15 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Pal B. et al. A single-cell RNA expression atlas of normal, preneoplastic and tumorigenic states in the human breast. EMBO J. 40, e107333 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Karaayvaz M. et al. Unravelling subclonal heterogeneity and aggressive disease states in TNBC through single-cell RNA-seq. Nat. Commun 9, 1–10 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Chen S. et al. Single-cell analysis reveals transcriptomic remodellings in distinct cell types that contribute to human prostate cancer progression. Nat. Cell Biol 23, 87–98 (2021). [DOI] [PubMed] [Google Scholar]
- 16.Dong B. et al. Single-cell analysis supports a luminal-neuroendocrine transdifferentiation in human prostate cancer. Communications Biology 3, 1–15 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Olalekan S, Xie B, Back R, Eckart H, & Basu A. Characterizing the tumor microenvironment of metastatic ovarian cancer by single-cell transcriptomics. Cell Rep. 35, 109165 (2021). [DOI] [PubMed] [Google Scholar]
- 18.Li H. et al. Reference component analysis of single-cell transcriptomes elucidates cellular heterogeneity in human colorectal tumors. Nat. Genet 49, 708–718 (2017). [DOI] [PubMed] [Google Scholar]
- 19.Baron M. et al. The Stress-Like Cancer Cell State Is a Consistent Component of Tumorigenesis. Cell Syst 11, 536–546.e7 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Kwart D. et al. Cancer cell-derived type I interferons instruct tumor monocyte polarization. Cell Rep. 41, (2022). [DOI] [PubMed] [Google Scholar]
- 21.Harris AL Hypoxia — a key regulatory factor in tumour growth. Nat. Rev. Cancer 2, 38–47 (2002). [DOI] [PubMed] [Google Scholar]
- 22.Tirosh I. et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189–196 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Jones MG, Yang D, & Weissman JS New Tools for Lineage Tracing in Cancer In Vivo. Annual Review of Cancer Biology 7(1): 111–129. (2023). [Google Scholar]
- 24.Marjanovic ND et al. Emergence of a High-Plasticity Cell State during Lung Cancer Evolution. Cancer Cell 38, 229–246.e13 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Hoek KS et al. In vivo Switching of Human Melanoma Cells between Proliferative and Invasive States. Cancer Res. 68, 650–656 (2008). [DOI] [PubMed] [Google Scholar]
- 26.Wei Y. et al. Single-cell analysis and functional characterization uncover the stem cell hierarchies and developmental origins of rhabdomyosarcoma. Nature Cancer 3, 961–975 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Youssef KK et al. Identification of the cell lineage at the origin of basal cell carcinoma. Nat. Cell Biol 12, 299–305 (2010). [DOI] [PubMed] [Google Scholar]
- 28.Van Keymeulen A. et al. Reactivation of multipotency by oncogenic PIK3CA induces breast tumour heterogeneity. Nature 525, 119–123 (2015). [DOI] [PubMed] [Google Scholar]
- 29.Chaligne R. et al. Epigenetic encoding, heritability and plasticity of glioma transcriptional cell states. Nat. Genet 53, 1469–1479 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Bittner M. et al. Molecular classification of cutaneous malignant melanoma by gene expression profiling. Nature 406, 536–540 (2000). [DOI] [PubMed] [Google Scholar]
- 31.Hoek KS et al. Metastatic potential of melanomas defined by specific gene expression profiles with no BRAF signature. Pigment Cell Res. 19, 290–302 (2006). [DOI] [PubMed] [Google Scholar]
- 32.Sensi M. et al. Human Cutaneous Melanomas Lacking MITF and Melanocyte Differentiation Antigens Express a Functional Axl Receptor Kinase. J. Invest. Dermatol 131, 2448–2457 (2011). [DOI] [PubMed] [Google Scholar]
- 33.Garraway LA et al. Integrative genomic analyses identify MITF as a lineage survival oncogene amplified in malignant melanoma. Nature 436, 117–122 (2005). [DOI] [PubMed] [Google Scholar]
- 34.Rambow F. et al. Toward Minimal Residual Disease-Directed Therapy in Melanoma. Cell 174, 843–855.e19 (2018). [DOI] [PubMed] [Google Scholar]
- 35.Karras P. et al. A cellular hierarchy in melanoma uncouples growth and metastasis. Nature 610, 190–198 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Pozniak J. et al. A TCF4/BRD4-dependent regulatory network confers cross-resistance to targeted and immune checkpoint therapy in melanoma. bioRxiv 2022.08.11.502598 (2022) doi: 10.1101/2022.08.11.502598. [DOI] [Google Scholar]
- 37.Baggiolini A. et al. Developmental chromatin programs determine oncogenic competence in melanoma. Science 373, eabc1048 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Bronner ME & LeDouarin NM Development and evolution of the neural crest: an overview. Dev. Biol 366, 2–9 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Laughney AM et al. Regenerative lineages and immune-mediated pruning in lung cancer metastasis. Nat. Med 26, 259–269 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Yang D. et al. Lineage tracing reveals the phylodynamics, plasticity, and paths of tumor evolution. Cell 185, 1905–1923.e25 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Meng X, Cui G. & Peng G. Lung development and regeneration: newly defined cell types and progenitor status. Cell Regen 12, 5 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Herriges M. & Morrisey EE Lung development: orchestrating the generation and regeneration of a complex organ. Development 141, 502–513 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Snyder EL et al. Nkx2–1 represses a latent gastric differentiation program in lung adenocarcinoma. Mol. Cell 50, 185–199 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Juul NH et al. KRAS(G12D) drives lepidic adenocarcinoma through stem-cell reprogramming. Nature 619, 860–867 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Zhang L. et al. Integrated single-cell RNA sequencing analysis reveals distinct cellular and transcriptional modules associated with survival in lung cancer. Signal Transduction and Targeted Therapy 7, 1–13 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Tirosh I. & Suvà ML Tackling the Many Facets of Glioblastoma Heterogeneity. Cell stem cell vol. 26 303–304 (2020). [DOI] [PubMed] [Google Scholar]
- 47.Couturier CP et al. Single-cell RNA-seq reveals that glioblastoma recapitulates a normal neurodevelopmental hierarchy. Nat. Commun 11, 1–19 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Huse JT & Holland EC Targeting brain cancer: advances in the molecular pathology of malignant glioma and medulloblastoma. Nat. Rev. Cancer 10, 319–331 (2010). [DOI] [PubMed] [Google Scholar]
- 49.Biserova K, Jakovlevs A, Uljanovs R. & Strumfa I. Cancer Stem Cells: Significance in Origin, Pathogenesis and Treatment of Glioblastoma. Cells 10, (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Alcantara Llaguno SR & Parada LF Cell of origin of glioma: biological and clinical implications. Br. J. Cancer 115, 1445–1450 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Liu C. et al. Mosaic Analysis with Double Markers Reveals Tumor Cell of Origin in Glioma. Cell 146, 209–221 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Friedmann-Morvinski D. et al. Dedifferentiation of neurons and astrocytes by oncogenes can induce gliomas in mice. Science 338, 1080–1084 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Moffitt RA et al. Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nat. Genet 47, 1168–1178 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Bailey P. et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature 531, 47–52 (2016). [DOI] [PubMed] [Google Scholar]
- 55.Collisson EA et al. Subtypes of pancreatic ductal adenocarcinoma and their differing responses to therapy. Nat. Med 17, 500–503 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Raghavan S. et al. Microenvironment drives cell state, plasticity, and drug response in pancreatic cancer. Cell 184, 6119–6137.e26 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Hwang WL et al. Single-nucleus and spatial transcriptome profiling of pancreatic cancer identifies multicellular dynamics associated with neoadjuvant treatment. Nat. Genet 54, 1178–1191 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Isaacson A, & Spagnoli FM Pancreatic cell fate specification: insights into developmental mechanisms and their application for lineage reprogramming. Curr. Opin. Genet. Dev 70, 32–39 (2021). [DOI] [PubMed] [Google Scholar]
- 59.Martens S. et al. Discovery and 3D imaging of a novel ΔNp63-expressing basal cell type in human pancreatic ducts with implications in disease. Gut 71, 2030–2042 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Backx E. et al. On the Origin of Pancreatic Cancer: Molecular Tumor Subtypes in Perspective of Exocrine Cell Plasticity. Cellular and Molecular Gastroenterology and Hepatology 13, 1243–1253 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Zhang Y. et al. Development and stem cells of the esophagus. Semin. Cell Dev. Biol 66, 25–35 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Bonnet D. & Dick JE Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat. Med 3, 730–737 (1997). [DOI] [PubMed] [Google Scholar]
- 63.Pollyea DA & Jordan CT Therapeutic targeting of acute myeloid leukemia stem cells. Blood 129, 1627–1635 (2017). [DOI] [PubMed] [Google Scholar]
- 64.Tenen DG Disruption of differentiation in human cancer: AML shows the way. Nat. Rev. Cancer 3, 89–101 (2003). [DOI] [PubMed] [Google Scholar]
- 65.van Galen P. et al. Single-Cell RNA-Seq Reveals AML Hierarchies Relevant to Disease Progression and Immunity. Cell 176, 1265–1281.e24 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Laurenti E. & Göttgens B. From haematopoietic stem cells to complex differentiation landscapes. Nature 553, 418–426 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Lasry A. et al. An inflammatory state remodels the immune microenvironment and improves risk stratification in acute myeloid leukemia. Nature Cancer 4, 27–42 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Levine JH et al. Data-Driven Phenotypic Dissection of AML Reveals Progenitor-like Cells that Correlate with Prognosis. Cell 162, 184–197 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Cook DP & Vanderhyden BC Context specificity of the EMT transcriptional response. Nat. Commun 11, 1–9 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Friedmann-Morvinski D. & Verma IM Dedifferentiation and reprogramming: origins of cancer stem cells. EMBO Rep. 15, 244–253 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Nieto MA, Huang RY, Jackson RA & Thiery JP EMT: 2016. Cell 166, 21–45 (2016). [DOI] [PubMed] [Google Scholar]
- 72.Cook DP & Vanderhyden BC Transcriptional census of epithelial-mesenchymal plasticity in cancer. Sci Adv 8, eabi7640 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Tyler M. & Tirosh I. Decoupling epithelial-mesenchymal transitions from stromal profiles by integrative expression analysis. Nat. Commun 12, 1–13 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Visvader JE Cells of origin in cancer. Nature 469, 314–322 (2011). [DOI] [PubMed] [Google Scholar]
- 75.Johnson L. et al. Somatic activation of the K-ras oncogene causes early onset lung cancer in mice. Nature 410, 1111–1116 (2001). [DOI] [PubMed] [Google Scholar]
- 76.Ferone G. et al. SOX2 Is the Determining Oncogenic Switch in Promoting Lung Squamous Cell Carcinoma from Different Cells of Origin. Cancer Cell 30, 519–532 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Atlasi Y. & Stunnenberg HG The interplay of epigenetic marks during stem cell differentiation and development. Nat. Rev. Genet 18, 643–658 (2017). [DOI] [PubMed] [Google Scholar]
- 78.Huang H-T et al. A network of epigenetic regulators guides developmental haematopoiesis in vivo. Nat. Cell Biol 15, 1516–1525 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Carter B. & Zhao K. The epigenetic basis of cellular heterogeneity. Nat. Rev. Genet 22, 235–250 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Sottoriva A. et al. A Big Bang model of human colorectal tumor growth. Nat. Genet 47, 209–216 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Flavahan WA, Gaskell E. & Bernstein BE Epigenetic plasticity and the hallmarks of cancer. Science 357, (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Sneeringer CJ et al. Coordinated activities of wild-type plus mutant EZH2 drive tumor-associated hypertrimethylation of lysine 27 on histone H3 (H3K27) in human B-cell lymphomas. Proc. Natl. Acad. Sci. U. S. A 107, 20980–20985 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Yap DB et al. Somatic mutations at EZH2 Y641 act dominantly through a mechanism of selectively altered PRC2 catalytic activity, to increase H3K27 trimethylation. Blood 117, 2451–2459 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Loukas I. et al. Selective advantage of epigenetically disrupted cancer cells via phenotypic inertia. Cancer Cell 41, 70–87.e14 (2023). [DOI] [PubMed] [Google Scholar]
- 85.Moubarak RS et al. The histone demethylase PHF8 regulates TGFβ signaling and promotes melanoma metastasis. Sci Adv 8, eabi7127 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Augert A. et al. Targeting NOTCH activation in small cell lung cancer through LSD1 inhibition. Sci. Signal 12, (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Terranova CJ et al. Reprogramming of bivalent chromatin states in NRAS mutant melanoma suggests PRC2 inhibition as a therapeutic strategy. Cell Rep. 36, 109410 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Hall AW et al. Bivalent Chromatin Domains in Glioblastoma Reveal a Subtype-Specific Signature of Glioma Stem Cells. Cancer Res. 78, 2463–2474 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Abatti LE et al. Epigenetic reprogramming of a distal developmental enhancer cluster drives SOX2 overexpression in breast and lung adenocarcinoma. Nucleic Acids Res. 51, 10109–10131 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Dick JE Stem cell concepts renew cancer research. Blood 112, 4793–4807 (2008). [DOI] [PubMed] [Google Scholar]
- 91.Ailles LE & Weissman IL Cancer stem cells in solid tumors. Curr. Opin. Biotechnol 18, 460–466 (2007). [DOI] [PubMed] [Google Scholar]
- 92.Ferrell JE Jr. Bistability, bifurcations, and Waddington’s epigenetic landscape. Curr. Biol 22, R458–66 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.de Sousa e Melo F. et al. A distinct role for Lgr5 stem cells in primary and metastatic colon cancer. Nature 543, 676–680 (2017). [DOI] [PubMed] [Google Scholar]
- 94.Clevers H. The cancer stem cell: premises, promises and challenges. Nat. Med 17, 313–319 (2011). [DOI] [PubMed] [Google Scholar]
- 95.Quintanal-Villalonga Á et al. Lineage plasticity in cancer: a shared pathway of therapeutic resistance. Nat. Rev. Clin. Oncol 17, 360–371 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Leonetti A. et al. Resistance mechanisms to osimertinib in EGFR-mutated non-small cell lung cancer. Br. J. Cancer 121, 725–737 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Quintanal-Villalonga A. et al. Multiomic Analysis of Lung Tumors Defines Pathways Activated in Neuroendocrine Transformation. Cancer Discov. 11, 3028–3047 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Awad MM et al. Acquired Resistance to KRAS Inhibition in Cancer. N. Engl. J. Med 384, 2382–2393 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Quintanal-Villalonga A. et al. Comprehensive molecular characterization of lung tumors implicates AKT and MYC signaling in adenocarcinoma to squamous cell transdifferentiation. J. Hematol. Oncol 14, 170 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Tong X. et al. Adeno-to-squamous transition drives resistance to KRAS inhibition in LKB1 mutant lung cancer. Cancer Cell 42, 413–428.e7 (2024). [DOI] [PubMed] [Google Scholar]
- 101.Zou M. et al. Transdifferentiation as a Mechanism of Treatment Resistance in a Mouse Model of Castration-Resistant Prostate Cancer. Cancer Discov. 7, 736–749 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Rebello RJ et al. Prostate cancer. Nature Reviews Disease Primers 7, 1–27 (2021). [DOI] [PubMed] [Google Scholar]
- 103.Arnold SJ & Robertson EJ Making a commitment: cell lineage allocation and axis patterning in the early mouse embryo. Nat. Rev. Mol. Cell Biol 10, 91–103 (2009). [DOI] [PubMed] [Google Scholar]
- 104.Waldrip WR et al. Smad2 Signaling in Extraembryonic Tissues Determines Anterior-Posterior Polarity of the Early Mouse Embryo. Cell 92, 797–808 (1998). [DOI] [PubMed] [Google Scholar]
- 105.Chazaud C. & Rossant J. Disruption of early proximodistal patterning and AVE formation in Apc mutants. Development 133, 3379–3387 (2006). [DOI] [PubMed] [Google Scholar]
- 106.Weatherbee SD, Carroll SB & Grenier JK From DNA to Diversity. (2001). [Google Scholar]
- 107.Peterson CM, Buckley C, Holley S. & Menias CO Teratomas: a multimodality review. Curr. Probl. Diagn. Radiol 41, 210–219 (2012). [DOI] [PubMed] [Google Scholar]
- 108.Greenwald AC et al. Integrative spatial analysis reveals a multi-layered organization of glioblastoma. bioRxiv 2023.07.06.547924 (2023) doi: 10.1101/2023.07.06.547924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Rao A, Barkley D, França GS & Yanai I. Exploring tissue architecture using spatial transcriptomics. Nature 596, 211–220 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Ji AL et al. Multimodal Analysis of Composition and Spatial Architecture in Human Squamous Cell Carcinoma. Cell 182, 497–514.e22 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Atanasova KR et al. Epigenetic Small-Molecule Screen for Inhibition and Reversal of Acinar Ductal Metaplasia in Mouse Pancreatic Organoids. bioRxiv 2023.11.27.567685 (2023) doi: 10.1101/2023.11.27.567685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Travaglini KJ et al. A molecular cell atlas of the human lung from single-cell RNA sequencing. Nature 587, 619–625 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Qiu C. et al. A single-cell time-lapse of mouse prenatal development from gastrula to birth. Nature (2024) doi: 10.1038/s41586-024-07069-w. [DOI] [PMC free article] [PubMed] [Google Scholar]