Abstract
In addition to immune cells and fibroblasts, the tumor microenvironment consists of extracellular matrix, which contains collagens, whose architecture and remodeling dictate cancer development and progression. Collagen receptors expressed by cancer cells sense signals generated by microenvironmental alterations in collagen state to regulate cell behavior and metabolism. Discoidin domain receptor (DDR)1 is a key sensor of collagen fiber state and composition that controls tumor cell metabolism and growth, response to therapy and patient survival. This review focuses on DDR1 to NRF2 signaling, its modulation of autophagy and macropinocytosis and role in cancer and other diseases. Elucidating the regulation DDR1 activity and expression under different pathophysiological conditions will facilitate the discovery of new therapeutics and treatments.
Keywords: DDR1, Collagen, Collagen receptors, Autophagy, Macropinocytosis, Cancer
Collagen receptors: ECM sensing and intercellular communication
Cancer research has gone from being cancer cell centric to a more inclusive and holistic view in which the cancer is studied together with the tumor microenvironment (TME), which also contains immune cells, endothelial cells, fibroblasts and the extracellular matrix (ECM) they produce [1,2]. Collagens (COLs) are the most abundant ECM proteins, and they play important structural roles that affect tumor rigidity and TME composition. New results show that COLs, which are classified as fibrillar or lamellar, also have critical regulatory functions that affect tumor growth and metabolism through interactions with different COL receptors expressed on cancer cells and other TME constituents [3,4]. These regulatory functions have raised interest in the therapeutic targeting of COL receptors [3,5], which include integrins, the discoidin domain receptors DDR1 and 2, OSCAR, GPVI, G6b-B, leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1), and the mannose family receptor uPARAP/Endo180 (MRC2). Amongst these molecules, DDR1 has received much recent attention due to its ability to control cancer cell metabolism in response to COL remodeling and ability to modulate ECM architecture and TME composition [4,6,7]. This review is focused on current progress in understanding COL-DDR1 interactions and downstream signaling.
The DDR family of COL-activated tyrosine kinases
The first DDR family members were the Dictyostelium discoideum lectin-like proteins or agglutinins discoidin I and II [8]. In addition to discoidins, DDR1 and 2 bear homology to transmembrane receptor tyrosine kinases (RTKs), a feature that facilitated their molecular cloning [9,10]. Whereas DDR1 has five isoforms (DDR1a-e) that are generated through alternative splicing of its kinase domain, DDR2 has only a single isoform [11–14] (Figure 1). DDR1a-c are kinase-active, whereas DDR1d-e are inactive kinases. DDR1 is mainly expressed on epithelial cells of different tissues, while DDR2 is found in mesenchymal cells including fibroblasts, myofibroblasts, smooth muscle cells, and chondrocytes [15]. Unlike typical RTKs, DDR1 and DDR2 are activated by microenvironmental COLs or their fragments rather than much less abundant growth factors. Using a series of overlapping triple-helical peptides, DDR1 and DDR2 were found to bind the GVMGFO (O=hydroxyproline) motif which is present only in fibrillar COLs, such as COL I-III [16,17]. Curiously, however, DDR1 can also be activated by non-fibrillar COLs, such as COL IV. Replacement of DDR2 amino acids with the corresponding DDR1 sequence at the periphery of the GVMGFO peptide-binding interface enables COL IV binding to DDR2 [17].
Figure 1. Schematic representation of the five DDR1 isoforms and the single DDR2 form.

DDR1a, 1b, 1c, and DDR2 are enzymatically active tyrosine kinase receptors. DDR1d and 1e are enzymatically inactive and their function is unknown. DS, discoidin domain; DS-like, discoidin-like domain; EJM, extracellular juxtamembrane region; TM, transmembrane segment; CJM, cytosolic juxtamembrane region; KD, kinase domain. AA, Amino Acid.
DDR1 and DDR2 share high degree of sequence conservation. Both are single-span transmembrane proteins, with 4 different domains: an extracellular region containing a N-terminal discoidin homology (DS) domain and a DS-like domain, a juxtamembrane domain (JM) that includes an extracellular JM domain and a cytosolic JM domain, and a C-terminal tyrosine kinase domain (Figure 1). Two surface-exposed loops (S52-T57 and R105-K112) and one amino acid (S175) within the DS domain are responsible for COL binding [18]. On binding of triple-helical COLs DDR1 undergoes trans-autophosphorylation between adjacent dimers, after it has formed densely packed clusters [19–21]. These findings also reveal that DDR1 is activated in a slow manner of hours rather than seconds as for other RTKs. How DDR1 kinase activity is regulated in vivo is an outstanding question. A new study performed in pancreatic ductal adenocarcinoma (PDAC) shows that DDR1 activity is regulated by the cleavage state of COL I. Whereas COL I that has been cleaved by matrix metalloproteinases (MMPs) promotes DDR1 autophosphorylation, full length COL I induces DDR1 proteasomal degradation [4]. This is consistent with previous studies showing that COL peptides have higher binding affinity to DDR1 than full length COLs [17] and DDR1 protein level is downregulated by COL fibrils [21]. Whether MMP-generated COL I fragments act similarly to small COL peptides and how full-length COL I promotes DDR1 degradation remains to be determined. The ability to differentially respond to full length COLs vs their MMP cleavage products sets DDR1 apart from other COL receptors and places it in a central position to control cancer metabolism in response to microenvironmental cues.
DDR1 and macroautophagy-macropinocytosis crosstalk
Macroautophagy (autophagy) is a conserved catabolic process in which denatured and aggregated proteins and organelles are encased within double membraned vesicles and are delivered to lysosomes for degradation [22–24]. Autophagy occurs via a series of well-defined steps: initiation, phagophore nucleation, elongation, autophagosome formation and autophagosome-lysosome fusion (Figure 2). Autophagy can be triggered by energy stress (nutrient deprivation) and AMPK activation or mTOR inhibition [22], playing important roles in health and disease. In cancer, autophagy is Janus-faced, being either tumor promoting or tumor inhibitory. A connection between autophagy and COL signaling is illustrated by glioblastoma stem cells whose homeostatic maintenance is regulated by COL I [25]. Moreover, DDR1 is the only COL receptor with kinase activity in glioblastoma and its inhibition induces therapy sensitizing autophagy [26]. 14–3-3 connects DDR1 to AKT which prevents Beclin-1 from initiating autophagy. Disruption of the DDR1:14–3-3:AKT:Beclin-1 complex impairs AKT and mTOR signaling and promotes formation of the Beclin-1:VPS34:ATG14 autophagy core complex (PI3K-III complex) (Figure 2 and Figure 3). DDR1 inhibition also alleviates osteoarthritis by blocking mTOR inhibition-induced autophagy.
Figure 2. A schematic overview of the (macro)autophagy pathway.

Under starvation conditions, autophagy is initiated by AMPK activation or mTOR inhibition, allowing ULK1 complex formation, and activation of the class III PtdIns3K (PI3K-III) complex which creates the PtdIns3P (PI3P)-rich regions (omegasome) on the surface of the ER membrane. The concerted action of ubiquitin-like conjugation system converts LC3-I to LC3-II, which leads to phagophore expansion or conversion of the isolation membrane into an autophagosome. Autophagosome and lysosome fusion is mediated by the SNARE complex result in formation autolysosomes within which the cargo is degraded [75]. The ULK1 complex, contains ULK1, ATG13, RB1CC1/FIP200 and ATG101; PI3K-III complex consists of ATG14, Beclin-1, PIK3R4/VPS15 and PIK3C3/VPS34; and the SNARE complex contains STX17, SNAP29, and VAMP8. Abbreviations: AMPK, AMP activated protein kinase; mTOR, mechanistic target of rapamycin kinase; ULK1, unc-51 like autophagy activating kinase 1; PtdIns3K, phosphatidylinositol 3-kinase; PI3P, phosphatidylinositol-3-phosphate; ER, endoplasmic reticulum; MAP1LC3/LC3, microtubule associated protein 1 light chain 3; RB1CC1/FIP200, retinoblastoma 1-inducible coiled-coil 1; PIK3R4/VPS15, phosphoinositide-3-kinase regulatory subunit 4; PIK3C3/VPS34, phosphatidylinositol 3-kinase catalytic subunit type 3; SNARE, soluble N-ethylmaleimide-sensitive factor (NSF) attachment protein receptor; STX17, syntaxin 17; SNAP29, soluble NSF attachment protein 29; VAMP8, vesicle-associated membrane protein 8; PE, phosphoethanolamine; ATG, autophagy-related protein.
Figure 3. Autophagy and macropinocytosis (MP) crosstalk.

On cCOL I binding, 14–3-3 connects activated DDR1 to AKT to prevent Beclin-1 from forming the Beclin-1:VPS34:VPS15:ATG14 autophagy core complex which promotes omegasome formation. Meanwhile, activated DDR1 promotes MP through the NF-κB-NRF2 module that induces transcription of MP-related genes. Loss of autophagy causes p62 accumulation which sequesters KEAP1 to activate NRF2-promoted MP.
In contrast to autophagy, macropinocytosis (MP) is an actin-dependent endocytic nutrient acquisition pathway in which soluble extracellular proteins are ingested and delivered to the lysosome in single membrane endocytic vesicles or macropinosomes [27]. Autophagy disruption causes accumulation of the autophagy chaperon SQSTM1/p62, which binds KEAP1 to induce transcription factor NRF2 activation [28], recently found to be the master activator of genes encoding MP-related proteins as well as MP activating growth factors and receptors [29]. DDR1 engagement by MMP-generated COL I fragments triggers NF-κB activation, SQSTM1/p62 induction and stimulation of NRF2-promoted MP [4], while suppressing autophagy through DDR1-Beclin-1 interaction [26]. Autophagy is normally inhibited when nutrients are high [22], but whether MP-produced ATP and amino acids inhibit autophagy needs to be determined. These results place DDR1 and NRF2 as key regulators of autophagy-MP crosstalk in those cancer cells that are surrounded by COL-rich stroma (Figure 3).
COL-DDR1 signaling in different cancers
PDAC
COL I-DDR1 signaling plays a critical role in PDAC metabolism and growth (Figure 4). DDR1 inhibition suppresses tumor growth and metastasis and improves the response to chemotherapy [4,30–32]. On activation, DDR1 directly interacts with focal adhesion kinase (FAK)-related protein tyrosine kinase 2 (PYK2), whose activation promotes cancer cell proliferation and metastasis through a p130 Crk-associated substrate scaffold (CAS)-RAP1-JNK1-c-Jun cascade [33]. DDR1-PYK2 activation also enhances expression of major ECM components [30]. Network-forming COL VIII promotes PDAC progression through DDR1 and integrin signaling, thereby activating pro-survival pathways, including PI3K-AKT and FAK-NF-κB [32]. MMP-mediated COL cleavage has been known to regulate tumor development [34] and a recent study showed that MMP-cleaved COL I (cCOL I) and intact COL I (iCOL I) differentially regulate PDAC bioenergetics, growth and metastatic spread via their effects on DDR1 activity and expression [4]. Whereas cCOL I engages DDR1 to activate an NF-κB-p62-NRF2-TFAM signaling cascade that stimulates MP, mitochondrial biogenesis, bioenergetics, and tumor growth, iCOL I triggers DDR1 proteasomal degradation, having opposite effects on these parameters. Patients with PDAC whose tumors are enriched for iCOL I and therefore are low DDR1 and NRF2 expressors exhibit greatly improved median survival compared to patients whose TME is high in MMP expression and enriched in cCOL I and their malignant cells are high in DDR1 and NRF2. These results justify development of therapeutics that target the DDR1-NF-κB-NRF2-TFAM metabolism simulating pathway. It’s also important to understand how DDR1, a rather weak RTK, exerts such profound effects on PDAC metabolism, although the answer may be somewhat trivial and related to the very high concentration of cCOL I in the TME, far surpassing the concentration of any growth factor by many orders of magnitude. cCOL I amounts in the PDAC TME correlate with high expression of inflammatory cytokines and MMPs [4]. cCOL I-DDR1 signaling may further augment TME inflammation by inducing cancer cell expression of CXCL5 via a PKCθ-SYK-NF-κB cascade, resulting in recruitment of tumor-associated neutrophils and formation of neutrophil extracellular traps [31]. In addition, tumor cell-derived α1(I) homotrimers resulting from epigenetic suppression of the COL1a2 gene promote PDAC development by activating DDR1 and α3β1 integrin on cancer cells and subsequent activation of FAK-PI3K-AKT signaling [35] (Figure 4). This was found to be associated with a microbiota enriched in anaerobic Bacteroidales in hypoxic and immunosuppressed tumors, and ablation of integrin β1 rather than DDR1 inhibits the autocrine response to COL I homotrimers. An earlier study showed that COL I homotrimers from lathyritic chick embryo tendons and calvaria have a greater solubility than COL I heterotrimers [36], providing a plausible explanation how the small amount of cancer cell derived COL I homotrimers may still lead to significant DDR1 activation [4].
Figure 4. COL-DDR1 signalings in cancer.

PDAC: (i) COL I activates DDR1-PYK2 and α2β1 integrin-FAK to stimulate cell proliferation and metastasis. Both receptor complexes rely on the p130 CAS-RAP1-JNK1-c-Jun cascade. (ii) COL VIII promotes PDAC progression through the DDR1-PI3K-AKT and integrin-FAK-NF-κB cascades. (iii) cCOL I engages DDR1 to activate an NF-κB-p62-NRF2-TFAM module that stimulates MP, mitochondrial biogenesis, bioenergetics, inflammation and tumor growth, iCOL I induces the opposite effects by causing the degradation of DDR1. (iv) cCOL I-DDR1 signaling modulates the TME by inducing CXCL5 expression via a PKCθ-SYK-NF-κB signaling cascade. (v) tumor cell-derived α1(I) homotrimers activate DDR1 and α3β1 integrin and inhibit tumor immunity via the FAK-PI3K-AKT pathway. TNBC: In a mouse model of TNBC, COL induced shedding of the DDR1 ectodomain (ECD) leads to alignment of wavy COL fibers resulting in prevention of T cell infiltration. However, in a human TNBC model (MDA-MB-231), COL III activated DDR1 maintains production of wavy COL III through activation of STAT1 to restrict cancer cell proliferation. In luminal BCa model, DDR1 inhibition promotes tumor growth through an unknown mechanism. HNSC: DDR1 promotes tumor growth in response to COL VIII and COL XI. Tumor-derived wavy COL III sustains tumor dormancy through DDR1-STAT1 signaling. HCC: cCOL I-activated DDR1 promotes tumor survival through the PI3K-AKT pathway and PSD4-ARF6-p38-ERK-c-Jun cascade. COL XV downregulates DDR1 expression through an unknown mechanism.
Breast cancer (BCa)
COL-DDR1 signaling supports BCa development by preventing effector T cell infiltration (Figure 4). BCa is the second most common cancer worldwide and is divided into several subtypes: luminal A, triple-negative BCa (TNBC), named after its lack of estrogen receptor alpha (ERα), progesterone receptor (PR) and epidermal growth factor receptor 2 (HER2), and HER2-positive BCa [37]. DDR1 is highly expressed in all subtypes compared to normal breast epithelium and DDR1 mutations are associated with poor prognosis in hormone receptor-positive patients [38]. The DDR1 kinase domain mutation R776W is supposed to be homologous to the EGFR hotspot mutation L858R [39]. In TNBC, DDR1 seems to promote tumor growth by inducing immune exclusion after cleavage of its COL binding ectodomain, which binds and aligns COL fibers [7]. However, how the DDR1 ectodomain aligns wavy COL fibers and how the aligned fibers prevent T cell invasion is unknown. Conversely, another study found that in an epithelial, human TNBC-derived MDA-MB-231 cell graft, DDR1 maintains a wavy form of tumor-derived COL III to restrict cancer cell proliferation through STAT1 activation [40] and that deletion of DDR1 in the MMTV-PyMT luminal BCa model converts the tumors to a more aggressive basal-type phenotype, resulting in increased lung metastasis through upregulation of fibrosis and hypoxia [41]. Although much more remains to be learned about DDR1 in BCa, its function seems to subtype and species specific.
Head and neck cancer (HNSCC)
DDR1 in HNSCC was also described as either stimulatory or inhibitory (Figure 4). Like BCa, DDR1 is overexpressed in HNSCC relative to normal tissue and shown to promote cancer cell growth and migration in response to COL VIII and XI [42]. However, tumor-derived COL III sustains tumor dormancy through DDR1-mediated STAT1 signaling, which tends to occur in lymph node-negative HNSCC that contain more COL III than lymph node-positive HNSCC [40]. The mechanism accounting for the differential signaling activities of COL VIII, XI and III is totally unknown, as DDR1 is still activated by other fibrillar COLs in the absence of COL I in PDAC [4].
Liver cancer
DDR1 is upregulated in mouse models of fibrosis-associated hepatocellular carcinoma (HCC) and corresponding patient specimens and high DDR1 is associated with poor prognosis [43,44] (Figure 4). As in PDAC, ECM containing cCOL I, but not iCOL I, activates DDR1 and downstream tumor-promoting effectors such as AKT [44]. DDR1 also interacts with ARF6 and activates it by recruiting PSD4, thereby stimulating MAPK signaling and metastasis [43]. Another interesting finding is that overexpression of COL XV, a basement membrane molecule that also interacts with DDR1, downregulates DDR1 mRNA and protein through an unknown mechanism [45–47]. Whether COL XV is a natural inhibitor of DDR1 that acts like iCOL I is an important question whose answer will provide new insights to DDR1 signaling. In contrast to its significant role in promoting HCC growth, DDR1 seems inessential in intrahepatic cholangiocarcinoma (ICC) that has been induced by hydrodynamic transduction of oncogenic drivers into hepatocytes [48]. Nonetheless, such results do not completely rule out DDR1’s role in ICC owing to the limitations of hydrodynamic transduction which mainly affects mature hepatocytes than hepatic stem cells or biliary epithelial cells which are also thought to give rise to ICC and can’t generate the desmoplastic microenvironment characteristic of human ICC [49,50].
COL-DDR signaling in neurodegenerative diseases
Although this review is focused on cancer, it should be mentioned that the ECM is also important for maintaining homeostasis in the central nervous system (CNS) and neuronal survival [51]. Structural, organizational, and functional ECM alterations due to genetic variation or environmental stress can result in neurodegenerative disease. As in cancer, different ECM components have opposing roles in neuropathology [52]. DDR1 and 2 are upregulated in Alzheimer’s and Parkinson’s diseases and their inhibition reduces α-synuclein, tau, and β-amyloid deposition and prevents cell loss, suggesting DDR1/2 inhibition as a potential target for clearance of neurotoxic proteins [53–55]. Differentially methylated regions (DMR) mapping in Parkinson’s patients and matched controls shows that eight CpG sites are hypomethylated in the DDR1 locus, suggesting that DDR1 as a molecular biomarker and a therapeutic target [56]. A tyrosine kinase inhibitor, nilotinib that potently inhibits DDR1, was found to induce autophagy and reduce inflammation and neurotoxicity [57], challenging the strategy of using COLs for induction of brain repair [58]. Another study reported that the cellular spread of alpha-synuclein fibrils depends on macropinocytic internalization and lysosomal degradation [59], implying an important role of autophagy-MP crosstalk in neurodegenerative diseases. Whether DDR1 promotes neurodegenerative disease through upregulation of MP and inhibition of autophagy and how DDR1 controls MP and autophagy in neurodegenerative diseases need to be further investigated. Anyhow, DDR1 is also an intriguing therapeutic target in neurodegenerative disease.
Concluding remarks
DDR1 is also tumor promoting in lung adenocarcinoma, retinoblastoma and colon carcinoma [60–62]. While the tumor promoting role of DDR1 can be easily explained through its effects on mitochondrial biogenesis, MP, and bioenergetics, how can a protein kinase that leads to NF-κB and NRF2 activation be tumor inhibitory in some cancers is not clear. This is certainly one of the major open questions regarding COL-DDR1 signaling (see Outstanding questions). A possible explanation could be divergent modes of COL remodeling and DDR1 gene mutations that alter substrate preference in different tumor contexts [63,64]. The targeting of DDR1 or DDR1 downstream effectors, including NF-κB, NRF2, mitochondrial biogenesis or MP may provide new opportunities for therapeutic development in those cancers whose metabolism and growth are DDR1 dependent (Table 1). DDR2 is also a promising target in PDAC [4,31,65], BCa [7,40,66–68], HNSCC [40,42,67], HCC [43,44,70,71] and ICC [48,72] (Table 1). Thus, dual DDR1 and DDR2 inhibitors may be quite effective. Indeed, several new DDR inhibitors found via high throughput screening for ATP competitive inhibitors or designed via in silico strategies have shown promising effects. KI-301690, a novel DDR1 inhibitor, potentiates the anticancer activity of gemcitabine in PDAC [30]. Another potent DDR1/2 inhibitor, VU6015929, with low cytotoxicity blocks COL-induced DDR1 activation and COL IV production, supporting the utility of DDR1/2 inhibition [73]. However, new and more selective and potent DDR inhibitors or combinations are needed to counteract drug resistance due to DDR1 and DDR2 mutations [64,74]. Inhibition of MP using tool compounds or blockade of mitochondrial protein synthesis with the FDA approved antibiotic tigecycline were also found to induce tumor regression in mouse PDAC models [4]. Whether and when any of these molecules or their derivatives will make it to the clinic is an open question.
Outstanding questions:
How does iCOL I induce DDR1 degradation?
How do non-fibrillar COLs, such as IV and XV bind DDR1 and how do they differentially control its activity or expression?
DDR1 regulates autophagy and MP through mTOR and NRF2, respectively. Can DDR1 directly phosphorylate autophagy- or MP-related proteins?
How does DDR1 activation in epithelial cancer cells affect tumor inflammation and immunity?
How can COL III, which is very similar to COL I, activate DDR1 and still have a diametrically opposing effect to COL I on tumor growth?
Can DDR2 substitute for DDR1? If yes, in what sense?
Which genes are specifically regulated by DDR1 or DDR2 and which genes can be regulated by both tyrosine kinases?
Are DDR1 and/or DDR2 involved in inside-out signaling and/or transactivation processes?
Table 1.
Overview of the functions of DDR1 and DDR2 in the development of distinct cancers.
| Cancer types | DDR1 function | DDR2 function |
|---|---|---|
| PDAC | Proliferation, metastasis and poor survival [4, 31] | Proliferation and migration activated by COL X [63] |
| BCa | Immune exclusion via promoting COL fiber alignment, or tumor dormancy though maintaining a wavy form of tumor-derived COL III [7, 40] | Proliferation, migration, invasion and immune exclusion [64–66] |
| HNSCC | Cell growth and migration in response to COLs III and XI or tumor dormancy in response to COL III [40, 42] | Migration, invasion [67] |
| HCC | Proliferation, metastasis and poor survival [43, 44] | EMT, metastasis, sorafenib resistance [68, 69] |
| ICC | Not essential in ICC growth [48] | Chinese patients had at least 1 actionable genetic aberration, with a significantly higher frequency in DDR2 compared with US patients [70] |
Highlights.
DDR1 is an important signal transducer that mediates crosstalk between cancer cells and their surrounding stroma.
Due to the very high concentrations of its ligand, cleaved collagen, DDR1 exerts a propound effect on tumor metabolism.
DDR1 controls macropinocytosis-autophagy crosstalk by activating NRF2, a key transcriptional regulator of metabolic adaptation.
Distinct collagen types and their cleavage products exert differential effects on DDR1 activity and expression.
DDR1 is an interesting therapeutic target for those cancers whose metabolism and growth depend on its activation.
Acknowledgments
We thank BioRender (BioRender.com) for plotting and acknowledge support by grants from the Scientific Research Starting Foundation of Affiliated Zhongshan Hospital of Fudan University (2023ZSQD03 to H.S.); the Scientific Research Starting Foundation of Fudan University (JIH1340033Y to H.S.); the General Program of the National Natural Science Foundation of China (82372884 to H.S.); the General Project of Shanghai Natural Science Foundation (23ZR1413600 to H.S.); Padres Pedal the Cause/C3 (PPTC2018 to M.K.); and the NIH (R01CA211794, R37AI043477, P01DK098108, U01CA274295 and U01AA027681 to M.K.); Additional support was provided by Ride the Point (M.K.); and the UC Pancreatic Cancer Consortium to M.K., who is the Ben and Wanda Hillyard Chair for Mitochondrial and Metabolic Diseases.
Footnotes
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.
Declaration of interests
The authors declare no conflicts of interest.
References:
- 1.Bejarano L et al. (2021) Therapeutic Targeting of the Tumor Microenvironment. Cancer Discov 11, 933–959 [DOI] [PubMed] [Google Scholar]
- 2.Sakemura R et al. (2022) Targeting cancer-associated fibroblasts in the bone marrow prevents resistance to CART-cell therapy in multiple myeloma. Blood 139, 3708–3721 [DOI] [PubMed] [Google Scholar]
- 3.Peng DH et al. (2020) Collagen promotes anti-PD-1/PD-L1 resistance in cancer through LAIR1-dependent CD8+ T cell exhaustion. Nat Commun 11, 4520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Su H et al. (2022) Collagenolysis-dependent DDR1 signalling dictates pancreatic cancer outcome. Nature 610, 366–372 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Ramos MIP et al. (2021) Cancer immunotherapy by NC410, a LAIR-2 Fc protein blocking human LAIR-collagen interaction. Elife 10, e62927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Elkamhawy A et al. (2021) The Journey of DDR1 and DDR2 Kinase Inhibitors as Rising Stars in the Fight Against Cancer. Int J Mol Sci 22, 6535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Sun X et al. (2021) Tumour DDR1 promotes collagen fibre alignment to instigate immune exclusion. Nature 599, 673–678 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Frazier WA et al. (1975) Purification and comparison of two developmentally regulated lectins from Dictyostelium discoideum. Discoidin I and II. J Biol Chem 250, 7714–7721 [PubMed] [Google Scholar]
- 9.Shrivastava A et al. (1997) An orphan receptor tyrosine kinase family whose members serve as nonintegrin collagen receptors. Mol Cell 1, 25–34 [DOI] [PubMed] [Google Scholar]
- 10.Vogel W et al. (1997) The discoidin domain receptor tyrosine kinases are activated by collagen. Mol Cell 1, 13–23 [DOI] [PubMed] [Google Scholar]
- 11.Alves F et al. (2001) Identification of two novel, kinase-deficient variants of discoidin domain receptor 1: differential expression in human colon cancer cell lines. FASEB J 15, 1321–1323 [DOI] [PubMed] [Google Scholar]
- 12.Alves F et al. (1995) Distinct structural characteristics of discoidin I subfamily receptor tyrosine kinases and complementary expression in human cancer. Oncogene 10, 609–618 [PubMed] [Google Scholar]
- 13.Perez JL et al. (1996) Identification of two isoforms of the Cak receptor kinase that are coexpressed in breast tumor cell lines. Oncogene 12, 1469–1477 [PubMed] [Google Scholar]
- 14.Karn T et al. (1993) Structure, expression and chromosomal mapping of TKT from man and mouse: a new subclass of receptor tyrosine kinases with a factor VIII-like domain. Oncogene 8, 3433–3440 [PubMed] [Google Scholar]
- 15.Theocharis AD et al. (2019) The extracellular matrix as a multitasking player in disease. FEBS J 286, 2830–2869 [DOI] [PubMed] [Google Scholar]
- 16.Konitsiotis AD et al. (2008) Characterization of high affinity binding motifs for the discoidin domain receptor DDR2 in collagen. J Biol Chem 283, 6861–6868 [DOI] [PubMed] [Google Scholar]
- 17.Xu H et al. (2011) Collagen binding specificity of the discoidin domain receptors: binding sites on collagens II and III and molecular determinants for collagen IV recognition by DDR1. Matrix Biol 30, 16–26 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Abdulhussein R et al. (2004) Exploring the collagen-binding site of the DDR1 tyrosine kinase receptor. J Biol Chem 279, 31462–31470 [DOI] [PubMed] [Google Scholar]
- 19.Juskaite V et al. (2017) Collagen induces activation of DDR1 through lateral dimer association and phosphorylation between dimers. eLife 6, e25716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Corcoran DS et al. (2019) DDR1 autophosphorylation is a result of aggregation into dense clusters. Sci Rep 9, 17104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Yeung DA et al. (2019) Clustering, Spatial Distribution, and Phosphorylation of Discoidin Domain Receptors 1 and 2 in Response to Soluble Collagen I. J Mol Biol 431, 368–390 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Dikic I and Elazar Z (2018) Mechanism and medical implications of mammalian autophagy. Nat Rev Mol Cell Biol 19, 349–364 [DOI] [PubMed] [Google Scholar]
- 23.Su H et al. (2017) VPS34 Acetylation Controls Its Lipid Kinase Activity and the Initiation of Canonical and Non-canonical Autophagy. Mol. Cell 67, 907–921.e7 [DOI] [PubMed] [Google Scholar]
- 24.Su H and Liu W (2018) PIK3C3/VPS34 control by acetylation. Autophagy 14, 1086–1087 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Motegi H et al. (2014) Type 1 collagen as a potential niche component for CD133-positive glioblastoma cells. Neuropathology 34, 378–385 [DOI] [PubMed] [Google Scholar]
- 26.Vehlow A et al. (2019) Interaction of Discoidin Domain Receptor 1 with a 14–3-3-Beclin-1-Akt1 Complex Modulates Glioblastoma Therapy Sensitivity. Cell Reports 26, 3672–3683.e7 [DOI] [PubMed] [Google Scholar]
- 27.Kay RR (2021) Macropinocytosis: Biology and mechanisms. Cells Dev 168, 203713. [DOI] [PubMed] [Google Scholar]
- 28.Ichimura Y and Komatsu M (2018) Activation of p62/SQSTM1-Keap1-Nuclear Factor Erythroid 2-Related Factor 2 Pathway in Cancer. Front Oncol 8, 210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Su H et al. (2021) Cancer cells escape autophagy inhibition via NRF2-induced macropinocytosis. Cancer Cell DOI: 10.1016/j.ccell.2021.02.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Ko S et al. (2022) A novel DDR1 inhibitor enhances the anticancer activity of gemcitabine in pancreatic cancer. Am J Cancer Res 12, 4326–4342 [PMC free article] [PubMed] [Google Scholar]
- 31.Deng J et al. (2021) DDR1-induced neutrophil extracellular traps drive pancreatic cancer metastasis. JCI Insight 6, 146133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Yan B et al. (2022) Tumor and stroma COL8A1 secretion induces autocrine and paracrine progression signaling in pancreatic ductal adenocarcinoma. Matrix Biol 114, 84–107 [DOI] [PubMed] [Google Scholar]
- 33.Shintani Y et al. (2008) Collagen I-mediated up-regulation of N-cadherin requires cooperative signals from integrins and discoidin domain receptor 1. J Cell Biol 180, 1277–1289 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Erkan M et al. (2008) The activated stroma index is a novel and independent prognostic marker in pancreatic ductal adenocarcinoma. Clin Gastroenterol Hepatol 6, 1155–1161 [DOI] [PubMed] [Google Scholar]
- 35.Chen Y et al. (2022) Oncogenic collagen I homotrimers from cancer cells bind to α3β1 integrin and impact tumor microbiome and immunity to promote pancreatic cancer. Cancer Cell 40, 818–834.e9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Jimenez SA et al. (1977) Identification of collagen alpha1(I) trimer in embryonic chick tendons and calvaria. Biochem Biophys Res Commun 78, 1354–1361 [DOI] [PubMed] [Google Scholar]
- 37.Harbeck N et al. (2019) Breast cancer. Nat Rev Dis Primers 5, 66. [DOI] [PubMed] [Google Scholar]
- 38.Griffith OL et al. (2018) The prognostic effects of somatic mutations in ER-positive breast cancer. Nat Commun 9, 3476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Li AR et al. (2008) EGFR mutations in lung adenocarcinomas: clinical testing experience and relationship to EGFR gene copy number and immunohistochemical expression. J Mol Diagn 10, 242–248 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Di Martino JS et al. (2022) A tumor-derived type III collagen-rich ECM niche regulates tumor cell dormancy. Nat Cancer 3, 90–107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Takai K et al. (2018) Discoidin domain receptor 1 (DDR1) ablation promotes tissue fibrosis and hypoxia to induce aggressive basal-like breast cancers. Genes Dev 32, 244–257 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Lai SL et al. (2019) Collagen Induces a More Proliferative, Migratory and Chemoresistant Phenotype in Head and Neck Cancer via DDR1. Cancers (Basel) 11, 1766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Zhang X et al. (2022) DDR1 promotes hepatocellular carcinoma metastasis through recruiting PSD4 to ARF6. Oncogene 41, 1821–1834 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Filliol A et al. (2022) Opposing roles of hepatic stellate cell subpopulations in hepatocarcinogenesis. Nature 610, 356–365 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Yao T et al. (2022) Collagen XV mediated the epithelial-mesenchymal transition to inhibit hepatocellular carcinoma metastasis. J Gastrointest Oncol 13, 2472–2484 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Cox TR (2021) The matrix in cancer. Nat Rev Cancer 21, 217–238 [DOI] [PubMed] [Google Scholar]
- 47.Clementz AG et al. (2013) Collagen XV Inhibits Epithelial to Mesenchymal Transition in Pancreatic Adenocarcinoma Cells. PLoS ONE 8, e72250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Affo S et al. (2021) Promotion of cholangiocarcinoma growth by diverse cancer-associated fibroblast subpopulations. Cancer Cell 39, 866–882.e11 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Chen X and Calvisi DF (2014) Hydrodynamic transfection for generation of novel mouse models for liver cancer research. Am J Pathol 184, 912–923 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Fabris L et al. (2019) The tumour microenvironment and immune milieu of cholangiocarcinoma. Liver Int 39 Suppl 1, 63–78 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Rike WA and Stern S (2023) Proteins and Transcriptional Dysregulation of the Brain Extracellular Matrix in Parkinson’s Disease: A Systematic Review. Int J Mol Sci 24, 7435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Sun Y et al. (2021) Role of the Extracellular Matrix in Alzheimer’s Disease. Front. Aging Neurosci 13, 707466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Hebron M et al. (2017) Discoidin domain receptor inhibition reduces neuropathology and attenuates inflammation in neurodegeneration models. J Neuroimmunol 311, 1–9 [DOI] [PubMed] [Google Scholar]
- 54.Fowler AJ et al. (2019) Multikinase Abl/DDR/Src Inhibition Produces Optimal Effects for Tyrosine Kinase Inhibition in Neurodegeneration. Drugs R D 19, 149–166 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Pagan FL et al. (2019) Pharmacokinetics and pharmacodynamics of a single dose Nilotinib in individuals with Parkinson’s disease. Pharmacol Res Perspect 7, e00470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Henderson AR et al. (2021) DNA Methylation and Expression Profiles of Whole Blood in Parkinson’s Disease. Front Genet 12, 640266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Anderson KE et al. (2022) Alteration of Autophagy and Glial Activity in Nilotinib-Treated Huntington’s Disease Patients. Metabolites 12, 1225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Ucar B (2021) Natural biomaterials in brain repair: A focus on collagen. Neurochemistry International 146, 105033. [DOI] [PubMed] [Google Scholar]
- 59.Bayati A et al. (2022) Rapid macropinocytic transfer of α-synuclein to lysosomes. Cell Rep 40, 111102. [DOI] [PubMed] [Google Scholar]
- 60.Zhu J et al. (2022) Discoidin domain receptor 1 promotes lung adenocarcinoma migration via the AKT/snail signaling axis. Mol Biol Rep 49, 7275–7286 [DOI] [PubMed] [Google Scholar]
- 61.Hong F-U et al. (2022) Tumor specifically internalizing peptide “HN-1”: Targeting the putative receptor retinoblastoma-regulated discoidin domain receptor 1 involved in metastasis. World J Clin Oncol 13, 323–338 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Romayor I et al. (2021) Tumor DDR1 deficiency reduces liver metastasis by colon carcinoma and impairs stromal reaction. Am J Physiol Gastrointest Liver Physiol 320, G1002–G1013 [DOI] [PubMed] [Google Scholar]
- 63.Su H and Karin M (2023) Collagen architecture and signaling orchestrate cancer development. Trends Cancer DOI: 10.1016/j.trecan.2023.06.002 [DOI] [PubMed] [Google Scholar]
- 64.Fathi Z et al. (2018) Distribution of KRAS, DDR2, and TP53 gene mutations in lung cancer: An analysis of Iranian patients. PLoS One 13, e0200633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Wen Z et al. (2022) COL10A1-DDR2 axis promotes the progression of pancreatic cancer by regulating MEK/ERK signal transduction. Front Oncol 12, 1049345. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 66.Lin C-C et al. (2021) DDR2 upregulation confers ferroptosis susceptibility of recurrent breast tumors through the Hippo pathway. Oncogene 40, 2018–2034 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Tu MM et al. (2019) Targeting DDR2 enhances tumor response to anti-PD-1 immunotherapy. Sci Adv 5, eaav2437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Wu C et al. (2022) Co-expression of DDR2 and IFITM1 promotes breast cancer cell proliferation, migration and invasion and inhibits apoptosis. J Cancer Res Clin Oncol 148, 3385–3398 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Xu J et al. (2014) Overexpression of DDR2 contributes to cell invasion and migration in head and neck squamous cell carcinoma. Cancer Biol Ther 15, 612–622 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Cai Y et al. (2022) LncRNA CEBPA-DT promotes liver cancer metastasis through DDR2/β-catenin activation via interacting with hnRNPC. J Exp Clin Cancer Res 41, 335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Liu Q-Q et al. (2021) Amplification of DDR2 mediates sorafenib resistance through NF-κB/c-Rel signaling in hepatocellular carcinoma. Cell Biol Int 45, 1906–1916 [DOI] [PubMed] [Google Scholar]
- 72.Cao J et al. (2020) Intrahepatic Cholangiocarcinoma: Genomic Heterogeneity Between Eastern and Western Patients. JCO Precis Oncol 4, PO.18.00414 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Jeffries DE et al. (2020) Discovery of VU6015929: A Selective Discoidin Domain Receptor 1/2 (DDR1/2) Inhibitor to Explore the Role of DDR1 in Antifibrotic Therapy. ACS Med Chem Lett 11, 29–33 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Shenoy GP et al. (2023) Discoidin domain receptor inhibitors as anticancer agents: A systematic review on recent development of DDRs inhibitors, their resistance and structure activity relationship. Bioorg Chem 130, 106215. [DOI] [PubMed] [Google Scholar]
- 75.Itakura E et al. (2012) The hairpin-type tail-anchored SNARE syntaxin 17 targets to autophagosomes for fusion with endosomes/lysosomes. Cell 151, 1256–1269 [DOI] [PubMed] [Google Scholar]
