Skip to main content

Nanobiotechnology augmented cancer stem cell guided management of cancer: liquid-biopsy, imaging, and treatment

Abstract

Cancer stem cells (CSCs) represent both a key driving force and therapeutic target of tumoral carcinogenesis, tumor evolution, progression, and recurrence. CSC-guided tumor diagnosis, treatment, and surveillance are strategically significant in improving cancer patients’ overall survival. Due to the heterogeneity and plasticity of CSCs, high sensitivity, specificity, and outstanding targeting are demanded for CSC detection and targeting. Nanobiotechnologies, including biosensors, nano-probes, contrast enhancers, and drug delivery systems, share identical features required. Implementing these techniques may facilitate the overall performance of CSC detection and targeting. In this review, we focus on some of the most recent advances in how nanobiotechnologies leverage the characteristics of CSC to optimize cancer diagnosis and treatment in liquid biopsy, clinical imaging, and CSC-guided nano-treatment. Specifically, how nanobiotechnologies leverage the attributes of CSC to maximize the detection of circulating tumor DNA, circulating tumor cells, and exosomes, to improve positron emission computed tomography and magnetic resonance imaging, and to enhance the therapeutic effects of cytotoxic therapy, photodynamic therapy, immunotherapy therapy, and radioimmunotherapy are reviewed.

Background

Cancer stem cells (CSCs) are a small group of cancer cells with stem-like properties. They have the capabilities of self-renewal, differentiation, and hence tumorigenicity. Compared to common cancer cells, CSCs are often in a quiescent state, express an elevated level of drug efflux pumps, have a higher capacity for DNA repair as well as anti-apoptosis, can cross-talk with tumor microenvironment to build up shields for immune escape and in turn remodeled by tumor microenvironment to undergo epithelial-mesenchymal transition. Due to these characteristics, CSCs are less vulnerable to chemotherapy and radiotherapy and, therefore, become one of the major causes of cancer recurrence and metastasis. Targeting CSC is a rational strategy to detect early-stage cancers, reverse chemotherapy and radiotherapy resistance, and prevent metastasis.

However, targeting CSC is challenging not only due to their innate characteristics, including rarity and resistance to chemotherapy and radiotherapy but also due to their heterogeneity and plasticity [1]. CSCs are a group of cells with high intertumoral and intratumoral heterogeneities. CSCs from different types of tumors may arise from tissue-specific or pluripotent stem cells. Within a given tumor, CSCs can exhibit heterogeneity in terms of their phenotypic characteristics, such as their expression of cell surface markers, their sensitivity to chemotherapy or radiation, and ability to form metastases. This heterogeneity can arise from genetic mutations or epigenetic modifications during tumor evolution. CSCs can also exhibit plasticity, meaning they can switch between stem cell and non-stem cell states or phenotypes in response to changes in their microenvironment [2].

Compared to conventional methods aiming at CSC-targeting, nanobiotechnology offers several advantages in cancer diagnosis, imaging, and treatment. For cancer diagnosis, nanomaterials can be engineered to bind specific surface cancer stem cell markers and trigger signal amplification. For imaging, nanomaterials can be used as contrast agents for more precise tumor detection. For cancer treatment, nanobiotechnology-engineered drug delivery systems have considerable advantages in targeted drug delivery, enhanced drug permeation and retention, reduced toxicity, multi-functionality, and personalization. Previous reviews have introduced how nanobiotechnologies exploited CSC characteristics, including cell-surface markers, metabolism, and microenvironment for biological imaging and chemotherapeutic enhancement [3, 4]. In this review, we focus on some of the most recent advances in how nanobiotechnologies leverage the characteristics of CSC to optimize cancer diagnosis and treatment, specifically in liquid biopsy, positron emission computed tomography (PET) and magnetic resonance imaging (MRI), cytotoxic therapy, photodynamic therapy, immunotherapy therapy and radioimmunotherapy (Fig. 1).

Fig. 1
figure 1

Leveraging characteristics of cancer stem cells for liquid biopsy, clinical imaging, and cancer treatment

Nanobiotechnology-optimized CSC-directed liquid biopsy

Liquid biopsy is a diagnostic test that involves the analysis of body fluids. In tumor diagnosis and monitoring, the test detects the presence of circulating tumor cells (CTCs), cell-free circulating tumor DNA (ctDNA), circulating tumor-derived exosome, and other biomarkers in the blood, which can provide valuable information about tumor volume and disease status. Compared to tissue biopsy, liquid biopsy is non-invasive, practically repeatable, and can provide more systematic information disregarding intra-tumor and inter-tumor heterogeneity. Real-time information from liquid biopsy may offer promising opportunities for cancer diagnosis and treatment monitor. However, several limitations hindered the clinical application of liquid biopsy: (1) super-low concentration of CTCs and ctDNA in early-stage disease and certain types of cancer; (2) demand for more specific markers; (3) high-cost and turnover time for sequencing. In this part, we mainly review the current statuses of CTC, ctDNA, and circulating tumor-derived exosome detection and how characteristics of CSC are exploited for nanobiotechnology-modified optimizations (Fig. 2).

Fig. 2
figure 2

Nanobiotechnology-modified circulating tumor cell, tumor DNA, and tumor-derived exosome detection

Circulating tumor cell

CTCs are cancer cells detached from primary or metastatic tumor lesions that enter the blood or lymphatic system to travel along the circulation system and potentially form new tumors. The clinical significance of CTCs lies in their potential use as a biomarker for cancer diagnosis, prognosis, and treatment response monitoring. Studies have shown that the number of CTCs in a patient’s blood is associated with disease progression and poor prognosis in various types of cancer, including breast, lung, colorectal, and prostate cancer [5, 6]. Additionally, CTCs can provide information about the genetic and molecular characteristics of the tumor, which can help guide treatment decisions. Therefore, CTC analysis is one of the most important paradigms of liquid biopsy.

CTCs are detected in various types of cancer, including breast cancer, NSCLC, colorectal cancer, lung cancer, and ovarian cancer. The common criteria to distinguish CTC from normal cells were cell diameter, shape, and molecular markers. (> 9 μm) and specific surface markers (CD45 negative to exclude leukocyte contamination). A series of platforms have been launched for CTC detection in clinical and research settings: CellSearch, the first and the only Food and Drug Administration (FDA) approved CTC test, detect CTCs in patients with breast, colorectal, and prostate cancer (EpCAM+, cytokeratin 8+, 18+ and/or 19+ and CD45); Epic Sciences capture CTCs and classify them based on molecular markers (EpCAM, cytokeratins, HER2, androgen receptors, etc.) and morphological features including size and shape. However, CTC detection is still a relatively new and developing technology. Current CTC tests may have some potential drawbacks, including limited sensitivity, lack of specificity, high cost, and limited availability.

CTCs and CSCs are two groups of cancer cells with crucial clinical significance sharing overlapping properties. They may both undergo epithelial-mesenchymal transition and enhanced trans-endothelial migration. Although a higher number of CTCs detected in the periphery blood was associated with a higher risk of metastasis, only a limited fraction of CTCs can successfully manage to initiate metastatic tumor lesions. Accumulated evidence strongly suggested the existence of CSCs among CTCs: CTC cell lines derived from chemotherapy-naive stage IV colorectal cancer patients maintained with non-serum suspension culture condition had the strong tumor-initiating capacity and were able to differentiate into main intestinal lineages both in vitro and in vivo [7]; compared to colorectal cancer xenograft-derived organoid, CTC-derived organoid of the same xenograft presented increased stem cell marker expression [8]; genetic linage tracing of stemness activation of murine glioblastoma (GBM) CTCs with native microenvironment presented nearly 100% of SOX2 activation, while less than 5% of primary tumor cells were SOX2 activated. Additionally, almost all murine GBM CTCs expressed Olig2, and 40% expressed CD133 [9]; CSC markers are detected on a fraction of CTCs in a variety of types of cancer, including breast cancer (CD44), colorectal cancer (LGR5), non-small cell lung cancer (NSCLC) and ovarian cancer (BMI1, CD133, ALDH1) [10,11,12].

To achieve circulating CTC detection, there were several challenges to overcome: the rarity of CTCs from a draw of blood, the innate heterogeneity of CTCs, and the lack of standardized CTC detection methods. Circulating CSCs are evolving as potential biomarkers in combination with CTCs for liquid biopsy. The combination of the CTC and CSC markers presented significant prognosis-predicting value in colorectal cancer [13]. Compared to CSC marker negative CTC, detection of CSC marker positive CTC was associated with metastases, increased risk of relapse and death in late-stage breast cancer [14, 15], NSCLC [16], prostate cancer [17], pancreatic cancer [18] and ovarian cancer [19]. The addition of CSC marker-directed cell sorting would increase the heterogeneity of CTC detected by unitary EpCAM-directed cell sorting, enhancing the clinical significance of CTC detected. Therefore, taking advantage of circulating CSC identification might boost the development of CTC detection in clinical settings. Similar challenges remain for circulating CSC detection due to their rarity, heterogeneity, and plasticity. The application of nanobiotechnology is apt to increase circulating CSC detection by enhancement of CSC capture using nanomaterials with appropriate charge, magnetism, or binding affinity to CSC-specific antigens.

A few nanobiotechnology-augmented CTC detection strategies based on CSC marker detection achieved outstanding performance in human sample testing. Gao et al. screened out and modified a CD44 targeting aptamer C24S for preparation of C24S conjugated magnetic nanoparticles, which greatly enhanced CD44 targeting specificity and enabled easy isolation of CD44+ circulating cells using magnet [20]. Kwizera et al. designed a gold-titanium coupling layer-coated electro-microfluidic chip with a linkage of heterogeneous CTC capture antibodies, including anti-CD44 and anti-Epcam. Compared to single marker-directed CTC capturing, a combined capture strategy of multiple subtypes of CTCs did not hamper the capture purity [21]. Their work provided a conceivable solution to improve the coverage of CTC and decrease the false negative rate due to CTC heterogeneity. Stergiopoulou et al. reported a long-term follow-up of 13 breast cancer patients who received comprehensive CTC monitoring, including measurements of EpCAM, phenotypic analysis, CSC marker analysis, PIK3CA and ESR1 mutations, and ESR1 methylation. According to their study, all patients with at least one positive marker relapsed, and all patients with negative markers stayed progression-free during follow-up [22]. Although the cohort examined by Stergiopoulou et al. was small and the CTC detection strategy applied was not integrated, a comprehensive CTC detection strategy consisting of multiple measurements might help to improve the detection rate of circulating CSC compromised by CSC plasticity.

Circulating tumor DNA

Cell-free DNAs were fragments from apoptotic cells that shed into body fluids. CtDNAs were cell-free DNAs from apoptotic or necrotic cancer cells. CtDNA analysis can detect tumor-associated features, including tumor-specific mutations, structural variants, copy number alterations, epigenetic features, microsatellite instability status, gene expression patterns, and tumor heterogeneity. CtDNA has several advantages as one of the paradigms of liquid biopsy: the amount of ctDNA was reported to be associated with tumor burden, which makes ctDNA testing a promising way to detect minimally residual disease; the genetic alteration of ctDNA represents tumor mutation burden and therefore could be used as a potential predictive biomarker for treatment response to immune checkpoint inhibitors; the sequential monitoring of specific somatic genetic mutations from the primary tumor through ctDNA sequencing may provide dynamic evidence for predictive treatment response to specific targeted therapies. In recent years, the FDA cleared several ctDNA-based tests, including EGFR, KRAS, BRAF, and EZH2 mutation analyses for customized targeted treatment planning. Although not yet cleared by the FDA, the device for Natera’s ctDNA tests was granted breakthrough device designation. These breakthroughs powerfully drove the development of novel methods and technologies to improve the sensitivity, specificity, and accuracy of ctDNA analysis.

DNA sequencing and amplification-based approaches are the mainstream and currently the gold-standard technology of un-informed and informed ctDNA detection. However, long turnover, the high cost of DNA sequencing, and the requirement of specialized pretreatment of samples for amplification tests limited clinical ctDNA application. For informed ctDNA testing, low-cost, point-of-care, and pretreatment-free technology is urgently needed. Nanobiotechnology has been implemented in several aspects of ctDNA detection to achieve amplification-free, super-sensitive, low-cost, and real-time ctDNA detection. The most promising strategies include fluidics-based electrochemical sensors, fluidics-based surface-enhanced Raman scattering (SERS), surface plasmon resonance (SPR), field-effect transistors (FETs), and Förster resonance energy transfer (FRET) based nano-sensors.

Electrochemical sensors are one of the most frequently studied solutions to ctDNA detection due to their low cost, simplicity, portability, and robustness. Constant endeavors boosted continuous optimizations of the performance of electrochemical sensors in ctDNA detection from multiple monogenic single point-mutation detection by peptide nucleic acids capture combined with Si3N4/Au/Si nanostructured microelectrodes [23] to multiple hotspot mutation detection via combinational probes [24], from double-strand DNA detection [25] to methylated DNA detection [26]. In this field, the low detection limit is pushed to 10 aM and even lower [27].

SERS is another powerful way to detect ctDNA at a single molecular level. Qi et al. developed a SERS probe using single-walled carbon nanotubes, which detected KRAS G12DM as low as 0.3 fM in an aliquot of 0.5 µl sample [28]. To fulfill multiplex detection, Yi et al. synthesized several SERS probes with remarkably different single-band Raman scattering signals, allowing a 10-plex biomarkers detection [29]. Cao et al. developed ultrasensitive SERS probes Cu2O@DTNB@hp3-1/Cu2O@DTNB@hp4-1 (PIK3CA E542K), Cu2O@4-ATP@hp3-2/Cu2O@4-ATP@ hp4-2 (TP53), Pd-AuNRs@DTNB@HP1-1 (BRAF V600E) and Pd-AuNRs@4-MBA@HP1-2 (KRAS G12V) with translational potential in diagnosis and follow-up of NSCLC patients. They employed Cu2O octahedra and Pd-Au nanorods for modification of SERS reporters to enhance field coupling, on-chip capillary pump for exemption of external pumps and rapid mixing, catalytic hairpin assembly for non-enzymatic signal amplification [30, 31].

FETs are relatively novel biosensors for ctDNA detection. Du et al. developed a silicon nanowire array field-effect transistor biosensor for PIK3CA E542K detection. The sensor achieved an ultralow detection limit of 10 aM and a good linearity under the ctDNA concentration range from 0.1 fM to 100 pM [32]. Pei et al. constructed a wearable self-healing patch consisting of a 3D printed eutectic gallium-indium circuit that can endure 100% strain and a FET sensor (iMethy) functionalized with 5-methylcytosine monoclonal antibody for specific methylated ctDNA capture and detection. The FET sensor can detect ctDNA as low as 0.1 fM in vitro, distinguishing tumor-bearing mice from healthy controls [33]. These findings suggested the translational potential of FET ctDNA biosensors as dynamic point-of-care ctDNA detection devices.

SPR is another frequently reported method of ctDNA detection based on real-time and quantitative information. Pioneering studies laid the foundation of gold nanoparticle-based plasmonic bio-sensors for detecting DNA sequences [34, 35]. Recent works aim at both DNA sequence variation and DNA methylation detection. Anh et al. developed a SERS-based nano-plasmonic biosensor for E542K and E545K mutations of PIK3CA and immunogold colloids for two methyl-cytosines in the promoter region with detection concentration of 200 fM [36]. Noemi et al. exploited a functional poly-L-lysine-based surface layer and plasmonic biosensor to detect the binding of MBD2 to methylated DNA. Their detection method had a high sensitivity (0.1 pg/mL, approximately 1 fM if the length of ctDNA is considered), specificity (99.6%), and accuracy (99.4%) for detecting methylated DNA in human plasma samples from healthy donors and colorectal cancer patients [37].

Rapid fingerprint single molecular detection of ctDNA through FRET-based nano-sensors was also feasible. Kunal et al. designed a DNA sensor consisting of fluorescent-tagged capture and query probes. The capture of mutated target ctDNA would shorten the physical distance between the capture and the query probe, thus triggering FRET. The FRET ctDNA probes could detect two biomarkers in 10 s with a detection limit of 3 fM and a mutant fraction as low as 0.0001%. However, single-molecule fluorescence microscopy, which is not commonly available in usual clinical settings, is required for FRET signal identification [38].

The underlying rationale for the identification of ctDNA of CSC from total ctDNA can be addressed in the following aspects: CSC is considered as one of the drivers of tumor initiation, progression, metastasis, and resistance to therapy hence positive ctDNA of CSC may provide a more accurate reflection of tumor burden, metastatic potential and prediction of treatment response; the detection of ctDNA from CSCs may allow for earlier detection of recurrence, timely treatment, and improved patients’ outcomes; CSCs’ competence of handling intrinsic and extrinsic hazards guarantees the longevity of the population to accumulate and preserve incremental genetic features; considered as one of the main driving forces of tumor evolution that give rise to phenotypic tumor heterogeneity, a pool of CSC was hypothesized as a representation of overall tumor heterogeneity that enables the capture of tumor heterogeneity [39,40,41].

Biosensors targeting CSC-derived ctDNA were studied for early diagnosis and disease surveillance. Ashok et al. designed nano-engineered plasmonic meta-sensors for real-time Raman scattering mapping of DNAs from glioblastoma cells, cell-line derived glioblastoma stem cells, tumor samples, and serum. The DNA data were then incorporated with machine learning for glioblastoma diagnosis. The addition of glioblastoma stem cell data to machine learning training significantly improved the sensitivity (83.3–93.3%) and specificity (75–100%) of the algorithm [42]. The same team also constructed a quantum superstructure to enhance SERS and mapped the SERS signal spectrum of DNA from breast, lung, and colorectal cancer cells, their cancer stem cell counterparts as well as corresponding tumor samples. With the Raman signals of cancer cells and cancer stem cell DNA as training data, ctDNA detection was achieved in raw plasma samples with 97% sensitivity and 83% specificity. With the addition of CSC DNA data, the algorithm distinguished lung cancer from mixed cancers with a sensitivity of 83.33% and 96.15%. With tumor DNA profile supplemented with the training, the algorithm further distinguished lung cancer from breast cancer and colorectal cancer, providing proof-of-concept evidence that tumor origin identification could be achieved through SERS-based liquid biopsy. CSC-derived ctDNA was an indispensable measurement [41].

Circulating tumor-derived exosome

Exosomes are nano-sized (30 to 50 nm) vesicles containing various types of biomolecules, including proteins, lipids, and nucleic acids released by cells for cell-cell communication. Exosomes participate in various physiological and pathological processes, including immune response, inflammation, cancer, and neurological disorders. Compared to CTC and ctDNA, tumor-derived exosomes had several advantages as a liquid biopsy detection alternative. Exosomes are much more abundant and stable than CTC and ctDNA in the blood, which potentiate sample acquisition. Unlike CTC and ctDNA, which were shed from bulk tumors at later stages or apoptotic tumor cells, exosomes were secreted by tumor cells throughout the tumor initiation and progression phase. However, there were still some limitations regarding exosome testing in liquid biopsy. The primary two were the lack of standardized exosome isolation, characterization, and quantification methods and the difficulty distinguishing tumor cell-derived exosomes from normal cell-derived exosomes. The most commonly used surface markers to identify exosomes were CD9, CD63, CD81, CD82, CD37, CD53, CD151, ALIX, and HSP70 [43]. However, exosomes were quite heterogeneous groups, and it was suggested that exosomes would acquire a fraction of surface markers from their cell of origin. For example, exosomes derived from dendritic cells may express CD86 and CD83, and exosomes from B cells may express CD19 and CD20 [44].

The biological characteristics, function, and destination of exosomes are different based on the cell of origin, cargos contained, and surface markers [45]. Compared to cancer cell-derived exosomes, CSC-derived exosomes had a unique role in the CSC plasticity remodeling [46]. They were more intimately involved in the promotion of cancer cellular epithelial-mesenchymal transition Field [47], therapeutic resistance Field [48], angiogenesis, and immune interaction [47]. Therefore, detecting CSC-derived exosomes might reflect cancer relapse, dynamic progression, therapeutic resistance, etc. Unlike non-cancerous tissue-derived exosomes and cancer cell-derived exosomes, CSC-derived exosomes contained cancer stem cell-specific surface markers, stemness-specific proteins, self-renewal regulatory microRNAs, and cancer stem cell-specific mutation signatures. Major pancreatic stem cell markers, including CD44v6, CD104, and CXCR4, were enriched in corresponding pancreatic stem cell-derived exosomes [48]. Stemness-specific protein Nanog was detected in exosomes from high-grade serous ovarian cancer patients [49]. Oncogenic KRAS and KRAS G13D carrying CD133+ microvesicles derived from colorectal cancer were proven to promote chemotherapy resistance [50]. These specific features of cancer stem cell-derived exosomes may serve as biomarkers for cancer-specific exosome isolation and improve sensitivity and specificity for cancer diagnosis and monitoring when combined with the current liquid biopsy strategy.

Rupa et al. exploited a machine learning algorithm in combination with SERS data to detect CSC-derived exosomes from blood samples. They collected exosomes from fibroblasts, MDAMB231 (breast cancer), H69AR(lung cancer), COLO205 (colorectal cancer), and their CSC counterparts. The samples were examined by SERS, the signal amplified by their nanosensor, and the data was used for artificial neural network training. After adjustment based on non-cancerous signals, SERS data of CSC-derived exosomes, containing crucial signals for cancer diagnosis, turned out to be different from cancer cell-derived ones. The machine learning algorithm was able to distinguish cancer samples from the non-cancer samples with 100% sensitivity and 100% specificity and identify tumor origin (breast cancer, sensitivity 73%, specificity 88%; colorectal cancer, sensitivity 42%, specificity 100%; lung cancer, sensitivity 100%, specificity 70%) [51].

Nanobiotechnology-optimized CSC-directed imaging

CSC-directed imaging is an indispensable technique that helps to identify the rare population of CSCs responsible for tumor initiation, progression, metastasis, therapeutic resistance, and recurrence. The prospect of clinical application of CSC imaging is extensive, not limited to understanding CSC evolution, tumor metastasis evaluation, treatment response monitoring, prognostic outcome prediction, and novel anti-tumor strategy development. According to the general characteristics of CSCs, two major strategies for CSC imaging were commonly exploited: targeting CSC markers, CSC-featured pathways, or metabolic activities. Theoretically, CSC imaging would allow for a thorough evaluation of systemic dynamics and heterogeneities of CSCs in different sites and microenvironments, which could exempt the patients from successive painful histologic biopsies. CSC imaging is still a developing field that faces numerous challenges and limitations.

Nanoscale materials and technologies engineered by nanobiotechnology facilitated biological and clinical purposes in many ways, including bioimaging. Target-specific nanocarriers precisely delivered fluorescent, magnetic, and radioactive contrast to the tissue of interest [52]. Modified nanocarriers and nanosensors could amplify rare and otherwise undetectable signals. Nanobiotechnology can thus enable more accurate and non-invasive monitoring of rare populations like CSCs. Since CSCs were generally identified through acknowledged CSC markers but not CSC-associated pathways, here, we mainly focused on reviewing the most acknowledged CSC marker-directed imaging and corresponding nanobiotechnology-driven strategies.

CD133

CD133 is a penta-span membrane glycoprotein and one of the most well-acknowledged CSC markers in various types of cancer. Preclinical and proof-of-concept studies on PET imaging probes for targeting CD133 tumor cells have been performed in glioma, prostate, lung, liver, colorectal, and many other tumor models. Namely, Gaedicke et al. reported noninvasive PET imaging of orthotopic CD133+ xenografts and patient-derived CD133+ CSCs by 64Cu-NOTA-AC133 mAb [53]. Glumac et al. conjugated PET probe 89Zr-HA10 IgG for CD133+ tumor imaging and validated its specificity using CD133+ and CD133 aggressive variant prostate cancer tumor-bearing mice. Hu et al. screened out a CD133-directed 64Cu PET tracer 64Cu CM-2 and tested its imaging performance and specificity in human hepatoma, melanoma, glioblastoma, breast cancer (Huh-7, Bowes, U87MG, MDA-MB231) and murine melanoma B16F10 allografts [54]. While a PET scan was unavailable, CD133+ tumor imaging could be achieved through specific CD133 contrast enhancement agents through MRI imaging. Chen et al. designed a CD133 antibody conjugated ultrasmall superparamagnetic iron oxide (USPIO) for a specific signal reduction in both FSE T2-weighted and merged scenarios. In addition to specific imaging of CD133+ HT29 subcutaneous xenografts, USPIO-CD133 Ab could also delineate drug-induced murine brain tumors [55].

A few studies tested the feasibility of CD133-directed testing as indicators for early tumor detection. Based on the findings that CD133 was explicitly expressed in small cell lung cancer (SCLC) but not in other types of lung cancer, such as adenocarcinoma and squamous cell carcinoma or adjacent healthy tissues, Kunihiro et al. proposed a strategy for early detection of SCLC that involved CD133 based PET imaging and autoantibody detection. They synthesized a CD133 targeting probe 89Zr-DFO-αCD133, which reached tumoral uptake of more than 40% of the injected dose in a xenograft mouse model of SCLC. Additionally, they found that αCD133 autoantibody could be found within one year pre-SCLC-diagnosis in 20/31 SCLC patients but not 55 controls consisting of colon, pancreas, and non-SCLC patients from the cardiovascular health study and the prostate, lung, colorectal, and ovarian screening trial [56]. Although the implementation of PET scan as a screening test was not feasible, constrained to cost, equipment availability, and complexity of the procedure, the findings of Kunihiro et al. allow us to posit a reasonable three-step strategy for early tumor detection: regular blood test as the initial screening, PET-scan for added evidence in high-risk patients and then surgical biopsy for diagnosis and treatment.

In addition to early tumor detection, treatment monitoring by CD133-directed imaging was also evaluated preclinically. Jung et al. found that celecoxib might have a CD133 modulation effect, so they synthesized a PET probe 89Zr-DFO-AC133.1, which targeted the glycosylated epitope of CD133, validated its CD133 targeted imaging specificity in human colon adenocarcinoma HT29 xenografts and tested six-day short-term CD133 level monitoring post-celecoxib-treatment [57]. Clinical evaluation of CD133-directed imaging had not been carried out in humans, possibly due to technique limitations and rationale deficiency. On one hand, there is currently no FDA-approved CD133 targeted therapy, which does not prioritize the need for CD133-directed imaging. On the other hand, the specificity, stability, and safety of current pre-clinical CD133-targeted PET tracers need further improvement. Several CD133 agents were under investigation in clinical trials, and hopefully, this result might boost the development and clinical translation of CD133-directed imaging.

CD44

CD44 is one of the earliest and most commonly acknowledged CSC markers intimately involved in tumor invasion and metastasis. Due to alternative splicing, the CD44 coding gene generated several different exon-containing variants, including CD44v3, CD44v4, CD44v6, and CD44v8-10, in addition to a standard form of CD44. According to some researchers, CD44 variants were also considered promising cancer biomarkers. The functional involvement of CD44 and its variants in tumor progression made them both CSC markers and therapeutic targets. Hence, CD44 targeted imaging could be used to select patients for CD44 targeted therapy, determine the dose for radioimmunotherapy, or monitor treatment response.

Since 2007, sustained efforts have been made to strive for the clinical translation of CD44-targeted PET imaging from lab studies. Several humanized anti-CD44 and anti-CD44 variant monoclonal antibodies including bivatuzumab and RG7356 or RO5429083 were selected for non-invasive imaging of CD44 + tumors and tested in phase 1 clinical trials of breast cancer (NCT02254005), head and neck neoplasms (NCT02254018), and metastatic or locally advanced solid tumors (NCT01358903) [58]. The pioneering clinical investigation of nuclide-labeled bivatuzumab-directed imaging of CD44 + tumors seemed halted due to the severe side effects during bivatuzumab treatment. Although the phase I dose escalation study of RG7356 was not interrupted by safety concerns, both specific and non-specific uptake of RG7356 in normal tissues limited the use of RG7356 in a fashion of antibody-drug conjugate [59]. Considering that smaller molecular weight might allow for higher tumor penetration capacity, Philipp et al. screened out a single-chain fragment variable with high affinity to CD44. They tested the ability of CD44-directed PET imaging in the form of bivalent antibodies labeled with 64Cu and 89Zr [60]. The probe 89Zr-DFO-scFv-Fc-CD44 demonstrated outstanding tumor uptake, which warrants the possibility of further clinical evaluation. However, up to now, relevant in-human evidence was still inadequate.

In addition to general CD44-directed imaging, both direct and indirect CD44 variant-directed imaging were also investigated. Haylock et al. constructed a bivalent CD44v6 targeting Fab antibody fragment linked through a self-dimerizing helix-turn helix motif named AbD19384 as the probe base, which was further labeled with 125I or 124I. 124I-AbD19384 showed superior tumor imaging ability than 18F-Fluoro-2-deoxy-D-glucose (18F-FDG) in CD44 positive breast cancer model MDA-MB-231 bearing mice. The authors’ primary aim was to test the strategy using bivalent Fabs to achieve in vivo imaging, so they did not focus on the overall performance of the CD44v6 imaging [61]. Since a splice variant of CD44 [62] was reported to interact with and stabilize xCT, which was responsible for uptake of a radiotracer (S)-4- (3- 18F-Fluoropropyl)-L-Glutamic Acid (18F-FSPG), the correlation between tumor uptake of 18F-FSPG and system xC/CD44 were examined in a few mini-cohorts of patients with NSCLC, breast cancer, hepatocellular carcinoma and prostate cancer [63,64,65]. According to these in-human data, tumor uptake of 18F-FSPG might be representative of redox status but not necessarily the status of CD44. Therefore, indirect imaging might help understand certain CD44-relevant mechanisms but is not readily available for clinical translation.

CXCR4

As a chemokine receptor, CXCR4 is intimately involved in cell survival, proliferation, and migration. It has been found to be associated with patients’ survival in various types of cancer, including leukemia, multiple myeloma, and breast carcinoma. CXCR4 was also engaged in support of CSC properties such as self-renewal, differentiation, and treatment resistance for certain types of cancer through endogenous maintenance and CSC-CSC niche crosstalk mediation. Therefore, CXCR4 is considered a biomarker for radioresistant CSCs and a potential therapeutic target.

Compared to other CSC markers, CXCR4 targeted imaging was one of the most extensively studied, and very few went on to clinical trials. A wide range of CXCR4 tracers has been developed and tested preclinically: 64Cu-AMD3100 [66], 18F-T140 [67], 64Cu-AMD3465 [68], 68Ga-labeled highly specific targeted contrast agent 68Ga-CPCR4-2 [69], 68Ga-DOTA-4-FBn-TN14003 [70], 18F-FP-labeled Ac-TC14012 [71], 68Ga-CCIC16, N-11C-Methyl-AMD3465 [72], 68Ga-pentixafor [73,74,75], 68Ga-NOTA-NFB [76], 18F-NOTA-T140 [77]. Among these miscellaneous tracers, the most noted tracer backbone was pentixafor, a synthetic peptide analogous to CXCL12 that specifically binds to CXCR4. Preclinical studies showed that pentixafor-based PET-tracer 68Ga-Pentixafor exhibited high tumor-to-background contrast and outstanding CXCR4 specificity. To date, 68Ga-Pentixafor has been evaluated in multiple types of hematologic and solid malignancies and demonstrated promising diagnostic and treatment assessment values in multiple myomas, small-cell lung cancer, and several other types of hematologic malignancies (Fig. 3). 68Ga-Pentixafor presented superior background contrast and lesion detection sensitivity than 18F-FDG in several types of hematological cancers. 68Ga-Pentixafor imaging has the potential for disease follow-up, treatment response prediction, and patient selection for the directed therapy [73,74,75, 78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99] (Table 1).

Fig. 3
figure 3

68Ga-Pentixafor-PET imaging of hematologic and solid cancer patients. Enhanced uptake of 68Ga-Pentixafor in bone marrow tissue in MM patient #26 and bone marrow lesions in MM patient #23 [79]. B Enhanced uptake of 68Ga-Pentixafor in bone marrow tissue in recurrent WM patient #5 and multiple lymph nodes of WM patient #11 [81]. C 68Ga-Pentixafor PET imaging and corresponding CE-T1w MRI imaging of recurrent CNSL patient #1 at time points of pre-treatment, completed chemotherapy (week 8), and completed radiotherapy (week 25) [85]. D 68Ga-Pentixafor PET/MRI imaging and HE staining of post-treatment MALT patients #2 and #9 [90]. E and F The average SUVmax and TBR of 68Ga-Pentixafor in solid cancer patients [87]. MM, multiple myeloma; WM, waldenström macroglobulinemia; CNSL, central nervous system lymphoma; NSCLC, non–small cell lung carcinoma; NEN, neuroendocrine neoplasm; AML, acute myeloid leukemia; CCC, cholangiocarcinoma; CLL, chronic lymphocytic leukemia; MZL, marginal zone lymphoma; DSRCT, desmoplastic small round cell tumor; SCLC, small cell lung carcinoma; ALL, acute lymphoblastoid leukemia. All panels were reproduced with permission

Table 1 68Ga-Pentixafor imaging in hematological and solid cancer patients

EpCAM

EpCAM was a classic epithelial cell marker ubiquitously expressed in epithelial cells and stem cells. It was once proposed as a CSC marker due to its tumor-promoting function and involvement in stemness regulating pathways, including Wnt/β-catenin and TGF-β/SMAD pathways [100]. Catumaxomab, a trifunctional bispecific antibody targeting EpCAM-expressing tumor cells, CD3-expressing T cells, and antigen-presenting cells, was approved for treating malignant ascites of patients with positive EpCAM. An initial attempt for PET imaging of EpCAM expressing tumors was achieved in A-431 tumor-bearing athymic mice using 68Ga-labelled HBED-CC scFv429, an HBED chelated diabody [101]. Recent researchers tested PET imaging of EpCAM in MDA-MB-231 tumors with a 64Cu−aptamer radiotracer [102]. Sergey et al. adopted a strategy in which a designed ankyrin repeat protein Ec1 was employed for specific EpCAM-binding. They evaluated the combination of two different positional labeling of Ec1 and four different radioactive nuclides 68Ga, 111In, 57Co, and 125I. According to their results, 125I-PIB-H6-Ec1 outcompeted all other tracers. It demonstrated the most outstanding tumor-to-background contrast as early as 3 to 6 h post-infection in tumor models of pancreatic cancer, ovarian cancer, and triple-negative breast cancer [103,104,105].

Nanobiotechnology-optimized CSC eradication

Due to the innate characteristics of CSCs, conventional therapies such as chemotherapy and radiotherapy that effectively targeted bulk tumor cells often failed to eradicate CSCs, which led to CSC enrichment. Moreover, CSC phenotypes were somewhat diversified by chemotherapeutic or radiotherapeutic modulation of both CSC and CSC niches. The post-conventional therapeutic enrichment and diversification of CSCs would lead to inevitable tumor relapse and metastasis. Therefore, eradicating CSCs might require combining strategies, including conventional therapy sensitization, CSC differentiation modulation, CSC niche targeting, and tumor immunity activation. Nanobiotechnology has several advantages in treating CSCs, including precise targeting, high-dose drug delivery, multiple drug delivery, controlled drug release, and artificial antigenicity [106]. Here, we reviewed recent advances in nanobiotechnology-optimized CSC-targeted treatment with in vivo validation in routine clinical cancer management including cytotoxic therapy, photodynamic therapy, immunotherapy, and radioimmunotherapy. Strategies regarding sonodynamic therapy and ablation therapy were formerly explicitly reviewed [107, 108].

Cytotoxic therapy

One of the major obstacles to CSC treatment was the innate resistance of CSC to conventional cytotoxic therapy. Several strategies were put forward: increase the drug dosage of specific CSC; modulation of resistance-associated genes or pathways; induction of CSC differentiation followed by conventional cytotoxic therapy; boosting various types of cellular death such as ferroptosis. However, specific CSC modulation requires precise delivery of modulators. To this end, many researchers studied the feasibility and anti-CSC efficacy of using CSC-targeted nanomaterial-based drug delivery systems. For CSC-specific targeted cytotoxic agents delivery, Ning et al. designed CD133 antibody conjugated nanoparticles to deliver SN-38, the active form of water-soluble camptothecin-11 (CD133Ab-NPs-SN-38) for CD133+ CSC targeting tested in HCT116 human colorectal cancer xenografts [109]. Li et al. synthesized HA-grafted all-trans-retinoic acid cationic nanoparticle (HA-eNPs/ATRA) and validated its anti-CSC/cancer effect in CD44 enriched B16F10 murine melanoma tumor model [110]. Wang et al. tested their doxorubicin-loaded HA-Lys-La nanoparticles (X-NP-DOX) in human breast cancer cell line MCF-7 bearing mice. They found significantly increased drug enrichment in the tumor. X-NP-DOX significantly reduced the tumorgenicity of CSC and exhibited an enhanced anti-tumor effect [111]. For CSC differentiation treatment, Geng et al. utilized a hypothesis-free method screening out nanoparticle C6NP, which has the most remarkable ability to induce CSC differentiation and sensitize CSC to conventional cytotoxic therapy [112]. A recent study by Wu et al. reported amplified ferroptosis through an iron oxyhydroxide-based nanosystem (FeOOH/siPROM2@HA) using a “three-pronged” strategy [113].

Photodynamic therapy

Photodynamic therapy exploits the reactive oxygen species generated by light-activated photosensitizers to eliminate cancer cells. Since FDA approved clinical photodynamic therapy in dermatological tumors, studies into photodynamic therapy aiming at effect enhancement and scope expansion have bloomed. Though compared to conventional therapy, precise targeting and controlled activation significantly reduced the systematic side-effects of photodynamic therapy, it faced limitations such as low tumor specificity and hypoxic tumor microenvironment. A combination of nano-drug delivery systems and photodynamic therapy might increase the overall systematic solubility, stability, target specificity, and biocompatibility and achieve synergetic effects through the co-delivery of photosensitizers and cytotoxic agents. The fundamental element of CSC targeting photodynamic therapy strategy usually includes a CSC targeting nanomaterial or CSC ligand, a photosensitizer, and sometimes added chemotherapeutic or CSC modulating agents. However, the cytotoxic effects of primordial photodynamic strategies against CSCs were far from satisfactory due to the original hypoxic state of CSCs and their high tolerance to reactive oxidative damage. A few studies focused on CSC targeting through nano-drug carrier-mediated photodynamic therapy and achieved particular progress.

One of the advantages of photodynamic therapy was that loaded drugs could be precisely released upon photo-activation. Ren et al. reported that by near-infrared activation of the sequential release of co-delivered miR-21 inhibitor and doxorubicin, breast cancer cells and CSCs were synergistically inhibited both in vitro and in vivo. At the same time, the synergetic effect of which was failed by simple co-delivery of the two drugs [114]. Exploiting photodynamic activation, Lee et al. constructed a CD44 targeting ROS-cleavable thioketal-SN38 conjugated hyaluronan-cholesterol nanoparticles. They successfully validated its drug delivery, drug release, and therapeutic effects using taxol-resistant ovarian cancer cell line HEY-30 bearing BALB/c [115]. Jung et al. developed a multi-functional CSC targeting system CA9-BPS-Cu(II), which combined chemo- and photodynamic effects with an acetazolamide-based approach [116]. In vitro, spheroid formation assay validated the cytotoxic effects of CA9-BPS-Cu(II) to CD133+ and CD44+/CD24 MDA-MB-231 breast cancer cells. The CSC marker ALDH and stemness-associated transcription factors such as OCT4, Nanog, SOX9, and Stat3 were significantly inhibited.

To cope with the hypoxic environment around CSC, Ning et al. designed a CD44 targeting type one aggregation-induced emission photosensitizer-loaded biomimetic mesoporous organosilicon nano-system to prevent cancer recurrence after radiotherapy [117]. Their nano-system achieved in vivo CSC targeting and significant tumor inhibition combined with radiotherapy. The proportion of in vivo CSC markers, including ALDH and CD133, significantly decreased. Considering the possible reduction of photo-responsiveness when conjugating photosensitizer directly to a specific antibody, Wang et al. proposed non-specific CSC targeting through a ribosome targeting [118]. In case CSCs were found to resist photodynamic therapy, novel strategies were investigated. Photochemical internalization, a highly efficient method of macromolecule delivery, especially for toxins that tended to be trapped in endosomes, had the potential to circumvent CSC photodynamic therapy resistance. Bostad et al. constructed a CD133 targeting photochemical internalization system PCIAC133−saponin and validated its CSC cytotoxicity in vitro and in vivo [114]. Multifunctional nanoplatforms with PET imaging ability and anti-cancer effect were extensively studied to fill the gap between simultaneous imaging and NIR treatment. Dong et al. designed a pH, HAse, and NIR responsive nano platform, MoS2-PEI-HA, which could be conjugated with either doxorubicin or 64Cu-NOTA. With 64Cu-NOTA labeled MoS2-PEI-HA, tumor uptake and dosages could be more specifically evaluated pre-treatment, and with the NIR responsiveness of DOX@ MoS2-PEI-HA, the release rate of DOX could reach up to 77.4% [119].

Immunotherapy

Immunotherapy mobilizes immune cells to attack cancer cells. Standard immunotherapy includes immune checkpoint inhibitors, immune system modulators, virus therapy, cancer vaccines, and engineered immune cell transfer. Despite high cancer targeting specificity and long-lasting effects in responsive patients, there were still challenges for immunotherapy to target CSC, which include heterogeneity, immune evasion, and microenvironment remodeling. The advantages of nanotechnology, including multiple drug delivery, multi-specific targeting, controlled drug release, and environmental responsiveness, can augment immunotherapy when dealing with these challenges for targeting CSC. Here, we systematically reviewed some up-to-date research on nanotechnology-assisted immunotherapeutic approaches targeting CSCs, including immune checkpoint inhibitors, cancer vaccines, and engineered immune cells (Fig. 4).

Fig. 4
figure 4

Schematic strategies and in vivo validation of nano-biotechnological optimized cancer stem cell-targeted immunotherapy. A Structural illustration of PEG-HA-OVA/PPLs. B and C Long-term in vivo validation of lung cancer protection by PEG-HA-OVA/PPLs [124]. D Graphical mechanism of vaccination against ALDH+ cancer stem cell. E and F Therapeutic effect of ALDH-ND against melanoma D5 tumor model [125]. G Schematic illustration of hydrogel application, CAR-MΦ conversion, cancer stem cell antigen presentation, adaptive immune response activation, and natural killing. H, I and J In vivo simulation of the post-surgical treatment effect of nano porter hydrogel [127]. HA, hyaluronic acid; OVA, ovalbumin; ND, nanodisc. All panels were reproduced with permission

Immune checkpoint inhibitors for cancer treatment have been one of the most active research areas of cancer immunotherapy. Advances in nanobiotechnology largely boosted the safety and efficacy of immune checkpoint inhibitors [120, 121]. While immune checkpoint inhibitors could eliminate the bulk tumor, CSCs demonstrated resistance to immune checkpoint inhibitors via expressing extremely high levels of immune checkpoint proteins, including PD-L1, CTLA-4, LAG-3, and TIM-3. However, administration of free immune checkpoint inhibitors would cause systematic adverse effects. Therefore, CSC-specific delivery of adequate immune checkpoint inhibitors would increase potency and reduce side effects. Several groups had focused on PD-L1 inhibitor delivery to CD44+ breast cancer cells. To supplement the anti-CSC effect in conventional chemo-/immunotherapy, Lang et al. developed a double-layered nanodevice loaded with PD-1/PD-L1 inhibitor HY19991 and anti-CSC agent thioridazine on the surface and paclitaxel inside. The nano delivery system delivers the loaded drugs to the tumor by passive targeting, and sequential release of the cargoes was triggered in response to the tumor microenvironment and endocytosis of the tumor cells [122]. The strategy of more recent studies to enhance PD-1/PD-L1 inhibitor potency on CD44+ CSC mostly involved (a) PD-1/PD-L1 inhibitor, (b) active targeting either through CD44 ligands including hyaluronic acid (HA) and chondroitin sulfate (CS), or anti-CD44 aptamer; (c) immunogenic antigen enhancement either by combination with chemotherapy or targeted therapy or implement of immunogenic foreign antigens. Kim et al. used HA as a CD44 targeting ligand to deliver PD-L1 siRNA for PD-L1 inhibition (PEG-HA-PPL) and ovalbumin (OVA) for immunogenic enhancement (PEG-HA-OVA) [123]. Cheng et al. equipped their paclitaxel/chloroquine delivery nanoparticle with CD44 targeting agent CS, immunogenic OVA, immunopotentiator CpG, and PD-L1 inhibitor atezolizumab (CpG + OVA + PTX + CQ-N/A). According to their results, PEG-HA-PPL/OVA, CpG + OVA + PTX + CQ-N/A, and all elicited tumor-reactive T cell-dependent anti-tumor effect and long-term protection in murine models [124].

However, to target CSC characterized by intracellular CSC markers such as ALDH, SOX2, and Nanog, direct biomarker-targeting through mAb was not feasible. In this case, immunogenic epitopes of corresponding CSC markers could be utilized to develop a specific therapeutic cancer vaccine. Alireza et al. developed a nanodisc (ND) vaccine delivery system to deliver antigenic ALDH1-A1 and ALDH1-A3 epitopes to antigen-presenting cells to induce specific ALDH+ CSC T cell response. They successfully constructed (ALDH-A1/A3-CpG)-ND of 9 to 13 nm, confirmed the lymphatic delivery and uptake of antigen in dendritic cells, B cells, and macrophages, validated T cell activation in vivo, and proved the robust therapeutic effects of (ALDH-A1/A3-CpG)-ND vaccine combined with anti-PD therapy in murine breast cancer models [125]. Their subsequent research identified four more SOX2 and Nanog epitopes with potential for vaccine development [126].

Adaptive chimeric antigen receptor (CAR) immune cell transfer is a more specific and potent way to target CSC. Artificially engineered CAR-expressing immune cells, including CAR-T, CAR-NK, CAR-DC, and CAR-M, could directly bind to cancer cells, significantly increasing cytotoxicity or antigen-presentation. Recent work by Chen et al. provided a simple way to induce local CD133-specific CAR-M through nanopore-hydrogel in post-surgical areas. They synthesized a CD68 promoter-driven anti-CD133 nanopore CAR (pCAR) and loaded to hydrogel coated with brain ECM-derived laminin/immune-stimulating peptides and citraconic anhydride–modified dextran to form macrophage-targeted NP-pCAR. NP-pCAR successfully induced anti-CD133 CAR-expressing macrophages, promoted phagocytosis of CD133+ cancer cells by CAR-M, activated T cell cytotoxicity, and elicited T cell memory [127].

Radioimmunotherapy

Radiopharmaceutical therapy is a type of cancer treatment that provides cancer-targeted radiation through the bloodstream via remedial radiopharmaceuticals. Radiopharmaceuticals consist of a radionuclide that emits radiation and a targeting ligand. Although radiopharmaceuticals were not as extensively used as other therapies such as surgery and chemotherapy, the number of radiopharmaceuticals designated for cancer diagnosis and treatment exceeds one-third of the current FDA-approved radiopharmaceuticals. In addition to the well-established application of iodine 131I-sodium iodide in carcinoma of thyroid and 223radium-dichloride in prostate cancer, more and more novel radiopharmaceuticals both for diagnosis and for treatment are getting approved by the FDA or undergoing clinical trials. Compared to conventional radiotherapy, radiopharmaceuticals have higher penetration capacity, can target primary and metastatic lesions in one shot, minimize average tissue exposure to radiation, and can even activate an anti-tumoral immune response when properly edited. Here, we mainly review how radiopharmaceuticals can be tailored to target CSCs.

CSCs represent only a tiny fraction of cancer cells and are resistant to chemotherapy, radiotherapy, and immunotherapy. Targeting CSCs through CSC-specific epitope-directed radionuclides is a promising therapeutic strategy. Several groups have tried to achieve high precision of CSC targeting based on current known CSC markers. In the past few years, 131I-antiAC133.1mAb, 131I-antiCD44mAb, 131I-antiCD44V6mAb, and 177Lu-antiCD44V6mAb were reported to have tumor inhibition effects in colorectal cancer HCT116 bearing xenografts [128,129,130]. However, since CD44 and CD133 were not solely expressed in CSC but also in many normal stem cells, the clinical validation of CD44 and CD133-directed therapy might meet foreseeable challenges. Until now, none of the CD44-directed radiopharmaceuticals has made it to clinical trial since the termination of two clinical trials of CD44v6 targeting antibody-drug conjugate bivatuzumab mertansine due to severe epidermal necrolysis.

Among the currently known CSC markers, CXCR4-directed radiopharmaceuticals achieved ground-breaking successes. In-human evaluation of CXCR4-directed radioligand therapy was performed in small-scale patients with multiple myoma [131, 132], advanced diffuse large B-cell lymphoma as part of the conditioning regimen before allogeneic stem cell transplantation [133], acute myeloid leukemia [134], advanced T-cell lymphoma [135]. The evaluated treatment strategy mainly involved selecting patients via 68Ga-Pentixafor imaging, pre-therapeutic dosimetry assessment through 177Lu-pentixather, and precision radioactive therapy by 90Y-pentixather. Almost all studies evaluating in-human 68Ga-Pentixafor imaging performance reported outstanding tolerance and zero major adverse events. However, regarding radiopharmaceutical therapy, non-negligible side effects were observed. A retrospective study assessed the side effects of radionuclide conjugated-pentixather in twenty-two patients with heavily pretreated hematopoietic cancers. The mild and slight probability of adverse events relating to drug elimination sites, including kidney and liver, added to the feasibility of pentixather-directed radiopharmaceutical therapy. However, hematopoietic side effects, the most frequent and serious ones, would limit pentixather-directed radiopharmaceutical therapy to hematopoietic stem cell transplantation setting [136]. With promising clinical translation pentixafor and pentixather, a novel generation of pentixafor and pentixather with higher specificity was studied. Replacement of the linker AMBS, which puts DOTA and CXCR4 binding core CPCR4 in pentixafor and iodoCPCR4 in pentixather together, by r-α-ABA would increase the hCXCR4 affinity of novel compounds tenfold. Although the novel 68GaDOTA-r-a-ABA-CPCR4 and -iodoCPCR4 did not show superior imaging performance than 68Ga-Pentixafor, the 48 h tumor-to-background ratios of 177Lu-DOTA-r-a-ABA-CPCR4 doubled even quadrupled compared to that of 177Lu-Pentixather [137]. The theranostic value of 177Lu-DOTA-r-a-ABA-CPCR4 as a second generation of CXCR4-directed radiopharmaceutical therapy was worth further evaluation.

Conclusion

CSCs represent both fundamental driving forces and therapeutic targets of tumoral carcinogenesis, tumor evolution, progression, and recurrence. Leveraging the characteristics of CSCs for early cancer diagnosis, dynamic tumor surveillance, and enhancement of therapeutic effects are facilitated by updating nanobiotechnologies. Though the field has accumulated fruitful results with clinical translational potential in the foreseeable future, tackling CSC remains challenging. The rarity, heterogeneity, and plasticity of CSCs call for ultrasensitive detection methods and multi-target targeting strategies. What we know about CSCs is still limited in varying genetic and epigenetic contexts of different cancer types. With an evolving understanding of the underlying mechanisms of maintenance of stem-like properties, the CSC niche, CSC-cancer evolution, the cross-talks between CSCs and tumor-microenvironment, novel markers, cellular targets, and cellular interaction modes can be exploited for holistic CSC targeting.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

CSC:

Cancer stem cell

CAR:

chimeric antigen receptor

CS:

chondroitin sulfate

CTC:

circulating tumor cell

ctDNA:

circulating tumor DNA

FDA:

Food and Drug Administration

FET:

field-effect transistor

FRET:

Förster resonance energy transfer

GBM:

glioblastoma

HA:

hyaluronic acid

MRI:

magnetic resonance imaging

ND:

nanodisc

NSCLC:

non-small cell lung cancer

OVA:

ovalbumin

PET:

positron emission computed tomography

SCLC:

small cell lung cancer

SERS:

surface enhanced Raman scattering

SPR:

surface plasmon resonance

USPIO:

ultrasmall superparamagnetic iron oxide

18F-FDG:

18F-Fluoro-2-deoxy-D-glucose

18F-FSPG:

(S)-4- (3- 18 F-Fluoropropyl)-L-Glutamic Acid

References

  1. Saygin C, Matei D, Majeti R, Reizes O, Lathia JD. Targeting Cancer Stemness in the clinic: from hype to Hope. Cell Stem Cell. 2019;24:25–40.

    Article  CAS  PubMed  Google Scholar 

  2. Batlle E, Clevers H. Cancer stem cells revisited. Nat Med. 2017;23:1124–34.

    Article  CAS  PubMed  Google Scholar 

  3. Han J, Won M, Kim JH, Jung E, Min K, Jangili P, Kim JS. Cancer stem cell-targeted bio-imaging and chemotherapeutic perspective. Chem Soc Rev. 2020;49:7856–78.

    Article  CAS  PubMed  Google Scholar 

  4. Zhang Z, Deng Q, Xiao C, Li Z, Yang X. Rational design of Nanotherapeutics based on the five features Principle for Potent Elimination of Cancer Stem cells. Acc Chem Res. 2022;55:526–36.

    Article  CAS  PubMed  Google Scholar 

  5. Cohen SJ, Punt CJ, Iannotti N, Saidman BH, Sabbath KD, Gabrail NY, Picus J, Morse M, Mitchell E, Miller MC, et al. Relationship of circulating tumor cells to tumor response, progression-free survival, and overall survival in patients with metastatic colorectal cancer. J Clin Oncol. 2008;26:3213–21.

    Article  PubMed  Google Scholar 

  6. Shimada R, Iinuma H, Akahane T, Horiuchi A, Watanabe T. Prognostic significance of CTCs and CSCs of tumor drainage vein blood in Dukes’ stage B and C colorectal cancer patients. Oncol Rep. 2012;27:947–53.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Grillet F, Bayet E, Villeronce O, Zappia L, Lagerqvist EL, Lunke S, Charafe-Jauffret E, Pham K, Molck C, Rolland N, et al. Circulating tumour cells from patients with colorectal cancer have cancer stem cell hallmarks in ex vivo culture. Gut. 2017;66:1802–10.

    Article  CAS  PubMed  Google Scholar 

  8. Le Du F, Fujii T, Kida K, Davis DW, Park M, Liu DD, Wu W, Chavez-MacGregor M, Barcenas CH, Valero V, et al. EpCAM-independent isolation of circulating tumor cells with epithelial-to-mesenchymal transition and cancer stem cell phenotypes using ApoStream(R) in patients with breast cancer treated with primary systemic therapy. PLoS ONE. 2020;15:e0229903.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Liu T, Xu H, Huang M, Ma W, Saxena D, Lustig RA, Alonso-Basanta M, Zhang Z, O’Rourke DM, Zhang L, et al. Circulating glioma cells exhibit Stem Cell-like Properties. Cancer Res. 2018;78:6632–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Koren A, Rijavec M, Kern I, Sodja E, Korosec P, Cufer T. BMI1, ALDH1A1, and CD133 Transcripts Connect Epithelial-Mesenchymal Transition to Cancer Stem Cells in Lung Carcinoma. Stem Cells Int 2016, 2016:9714315.

  11. Aktas B, Tewes M, Fehm T, Hauch S, Kimmig R, Kasimir-Bauer S. Stem cell and epithelial-mesenchymal transition markers are frequently overexpressed in circulating tumor cells of metastatic breast cancer patients. Breast Cancer Res. 2009;11:R46.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Blassl C, Kuhlmann JD, Webers A, Wimberger P, Fehm T, Neubauer H. Gene expression profiling of single circulating tumor cells in ovarian cancer - establishment of a multi-marker gene panel. Mol Oncol. 2016;10:1030–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Iinuma H, Watanabe T, Mimori K, Adachi M, Hayashi N, Tamura J, Matsuda K, Fukushima R, Okinaga K, Sasako M, Mori M. Clinical significance of circulating tumor cells, including cancer stem-like cells, in peripheral blood for recurrence and prognosis in patients with dukes’ stage B and C colorectal cancer. J Clin Oncol. 2011;29:1547–55.

    Article  PubMed  Google Scholar 

  14. Papadaki MA, Stoupis G, Theodoropoulos PA, Mavroudis D, Georgoulias V, Agelaki S. Circulating Tumor cells with stemness and Epithelial-To-Mesenchymal Transition Features Are Chemoresistant and Predictive of poor outcome in metastatic breast Cancer. Mol Cancer Ther. 2019;18:437–47.

    Article  CAS  PubMed  Google Scholar 

  15. Lee CH, Hsieh JC, Wu TM, Yeh TS, Wang HM, Lin YC, Chen JS, Lee CL, Huang WK, Hung TM, et al. Baseline circulating stem-like cells predict survival in patients with metastatic breast Cancer. BMC Cancer. 2019;19:1167.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Nel I, Jehn U, Gauler T, Hoffmann AC. Individual profiling of circulating tumor cell composition in patients with non-small cell lung cancer receiving platinum based treatment. Transl Lung Cancer Res. 2014;3:100–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Kerr BA, Miocinovic R, Smith AK, West XZ, Watts KE, Alzayed AW, Klink JC, Mir MC, Sturey T, Hansel DE, et al. CD117(+) cells in the circulation are predictive of advanced prostate cancer. Oncotarget. 2015;6:1889–97.

    Article  PubMed  Google Scholar 

  18. Poruk KE, Blackford AL, Weiss MJ, Cameron JL, He J, Goggins M, Rasheed ZA, Wolfgang CL, Wood LD. Circulating Tumor cells expressing markers of tumor-initiating cells predict poor survival and Cancer recurrence in patients with pancreatic ductal adenocarcinoma. Clin Cancer Res. 2017;23:2681–90.

    Article  CAS  PubMed  Google Scholar 

  19. Gening SO, Abakumova TV, Gafurbaeva DU, Rizvanov AA, Antoneeva II, Miftakhova RR, Peskov AB, Gening TP. The detection of Stem-Like circulating Tumor cells could increase the clinical applicability of Liquid Biopsy in Ovarian Cancer. Life (Basel) 2021, 11.

  20. Gao T, Li W, Ma J, Chen Y, Wang Z, Sun N, Pei R. Selection of DNA aptamer recognizing CD44 for high-e ffi ciency capture of circulating tumor cells. Talanta. 2023;262:124728.

    Article  CAS  PubMed  Google Scholar 

  21. Kwizera EA, Ou W, Lee S, Stewart S, Shamul JG, Xu J, Tait N, Tkaczuk KHR, He X. Greatly enhanced CTC Culture enabled by capturing CTC heterogeneity using a PEGylated PDMS-Titanium-Gold Electromicrofluidic device with glutathione-controlled Gentle Cell Release. ACS Nano. 2022;16:11374–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Stergiopoulou D, Markou A, Strati A, Zavridou M, Tzanikou E, Mastoraki S, Kallergi G, Georgoulias V, Lianidou E. Comprehensive liquid biopsy analysis as a tool for the early detection of minimal residual disease in breast cancer. Sci Rep. 2023;13:1258.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Das J, Ivanov I, Montermini L, Rak J, Sargent EH, Kelley SO. An electrochemical clamp assay for direct, rapid analysis of circulating nucleic acids in serum. Nat Chem. 2015;7:569–75.

    Article  CAS  PubMed  Google Scholar 

  24. Das J, Ivanov I, Safaei TS, Sargent EH, Kelley SO. Combinatorial probes for high-throughput Electrochemical Analysis of circulating nucleic acids in clinical samples. Angew Chem Int Ed Engl. 2018;57:3711–6.

    Article  CAS  PubMed  Google Scholar 

  25. Das J, Ivanov I, Sargent EH, Kelley SO. DNA Clutch Probes for circulating tumor DNA analysis. J Am Chem Soc. 2016;138:11009–16.

    Article  CAS  PubMed  Google Scholar 

  26. Povedano E, Montiel VR, Valverde A, Navarro-Villoslada F, Yanez-Sedeno P, Pedrero M, Montero-Calle A, Barderas R, Pelaez-Garcia A, Mendiola M, et al. Versatile Electroanalytical Bioplatforms for simultaneous determination of Cancer-related DNA 5-Methyl- and 5-Hydroxymethyl-cytosines at global and gene-specific levels in human serum and tissues. ACS Sens. 2019;4:227–34.

    Article  CAS  PubMed  Google Scholar 

  27. Xu XW, Weng XH, Wang CL, Lin WW, Liu AL, Chen W, Lin XH. Detection EGFR exon 19 status of lung cancer patients by DNA electrochemical biosensor. Biosens Bioelectron. 2016;80:411–7.

    Article  CAS  PubMed  Google Scholar 

  28. Zhou Q, Zheng J, Qing Z, Zheng M, Yang J, Yang S, Ying L, Yang R. Detection of circulating Tumor DNA in human blood via DNA-Mediated surface-enhanced Raman Spectroscopy of single-walled Carbon nanotubes. Anal Chem. 2016;88:4759–65.

    Article  CAS  PubMed  Google Scholar 

  29. Zeng Y, Ren JQ, Shen AG, Hu JM. Splicing nanoparticles-based click SERS could Aid Multiplex Liquid Biopsy and Accurate Cellular Imaging. J Am Chem Soc. 2018;140:10649–52.

    Article  CAS  PubMed  Google Scholar 

  30. Cao X, Ge S, Hua W, Zhou X, Lu W, Gu Y, Li Z, Qian Y. A pump-free and high-throughput microfluidic chip for highly sensitive SERS assay of gastric cancer-related circulating tumor DNA via a cascade signal amplification strategy. J Nanobiotechnol. 2022;20:271.

    Article  CAS  Google Scholar 

  31. Cao X, Ge S, Zhou X, Mao Y, Sun Y, Lu W, Ran M. A dual-signal amplification strategy based on pump-free SERS microfluidic chip for rapid and ultrasensitive detection of non-small cell lung cancer-related circulating tumour DNA in mice serum. Biosens Bioelectron. 2022;205:114110.

    Article  CAS  PubMed  Google Scholar 

  32. Li D, Chen H, Fan K, Labunov V, Lazarouk S, Yue X, Liu C, Yang X, Dong L, Wang G. A supersensitive silicon nanowire array biosensor for quantitating tumor marker ctDNA. Biosens Bioelectron. 2021;181:113147.

    Article  CAS  PubMed  Google Scholar 

  33. Fang P, Ji X, Zhao X, Yan-Do R, Wan Y, Wang Y, Zhang Y, Shi P. Self-Healing Electronics for Prognostic Monitoring of Methylated circulating Tumor DNAs. Adv Mater. 2023;35:e2207282.

    Article  PubMed  Google Scholar 

  34. Willets KA, Van Duyne RP. Localized surface plasmon resonance spectroscopy and sensing. Annu Rev Phys Chem. 2007;58:267–97.

    Article  CAS  PubMed  Google Scholar 

  35. Ma W, Kuang H, Xu L, Ding L, Xu C, Wang L, Kotov NA. Attomolar DNA detection with chiral nanorod assemblies. Nat Commun. 2013;4:2689.

    Article  PubMed  Google Scholar 

  36. Nguyen AH, Sim SJ. Nanoplasmonic biosensor: detection and amplification of dual bio-signatures of circulating tumor DNA. Biosens Bioelectron. 2015;67:443–9.

    Article  CAS  PubMed  Google Scholar 

  37. Bellassai N, D’Agata R, Marti A, Rozzi A, Volpi S, Allegretti M, Corradini R, Giacomini P, Huskens J, Spoto G. Detection of Tumor DNA in human plasma with a functional PLL-Based surface layer and Plasmonic Biosensing. ACS Sens. 2021;6:2307–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Khanna K, Mandal S, Blanchard AT, Tewari M, Johnson-Buck A, Walter NG. Rapid kinetic fingerprinting of single nucleic acid molecules by a FRET-based dynamic nanosensor. Biosens Bioelectron. 2021;190:113433.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Liu ZB, Ezzedine NE, Eterovic AK, Ensor JE, Huang HJ, Albanell J, Choi DS, Lluch A, Liu Y, Rojo F, et al. Detection of breast cancer stem cell gene mutations in circulating free DNA during the evolution of metastases. Breast Cancer Res Treat. 2019;178:251–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Abad E, Graifer D, Lyakhovich A. DNA damage response and resistance of cancer stem cells. Cancer Lett. 2020;474:106–17.

    Article  CAS  PubMed  Google Scholar 

  41. Haldavnekar R, Ganesh S, Venkatakrishnan K, Tan B. Cancer Stem Cell DNA enabled real-time genotyping with self-functionalized Quantum superstructures-overcoming the barriers of Noninvasive cfDNA Cancer Diagnostics. Small Methods. 2022;6:e2101467.

    Article  PubMed  Google Scholar 

  42. Dhinakaran AK, Ganesh S, Haldavnekar R, Tan B, Das S, Venkatakrishnan K. Holistic Analysis of Glioblastoma Stem cell DNA using Nanoengineered Plasmonic Metasensor for Glioblastoma Diagnosis. Small Methods. 2022;6:e2200547.

    Article  PubMed  Google Scholar 

  43. Kowal J, Arras G, Colombo M, Jouve M, Morath JP, Primdal-Bengtson B, Dingli F, Loew D, Tkach M, Thery C. Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proc Natl Acad Sci U S A. 2016;113:E968–977.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Guedes VA, Devoto C, Leete J, Sass D, Acott JD, Mithani S, Gill JM. Extracellular vesicle proteins and MicroRNAs as biomarkers for traumatic brain Injury. Front Neurol. 2020;11:663.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Grigoryeva ES, Tashireva LA, Savelieva OE, Zavyalova MV, Popova NO, Kuznetsov GA, Andryuhova ES, Perelmuter VM. The Association of Integrins beta3, beta4, and alphaVbeta5 on exosomes, CTCs and Tumor cells with localization of distant metastasis in breast Cancer patients. Int J Mol Sci 2023, 24.

  46. Li W, Zhang L, Guo B, Deng J, Wu S, Li F, Wang Y, Lu J, Zhou Y. Exosomal FMR1-AS1 facilitates maintaining cancer stem-like cell dynamic equilibrium via TLR7/NFkappaB/c-Myc signaling in female esophageal carcinoma. Mol Cancer. 2019;18:22.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Hwang WL, Lan HY, Cheng WC, Huang SC, Yang MH. Tumor stem-like cell-derived exosomal RNAs prime neutrophils for facilitating tumorigenesis of colon cancer. J Hematol Oncol. 2019;12:10.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Wang H, Rana S, Giese N, Buchler MW, Zoller M. Tspan8, CD44v6 and alpha6beta4 are biomarkers of migrating pancreatic cancer-initiating cells. Int J Cancer. 2013;133:416–26.

    Article  CAS  PubMed  Google Scholar 

  49. Sherman-Samis M, Onallah H, Holth A, Reich R, Davidson B. SOX2 and SOX9 are markers of clinically aggressive disease in metastatic high-grade serous carcinoma. Gynecol Oncol. 2019;153:651–60.

    Article  CAS  PubMed  Google Scholar 

  50. Kang M, Kim S, Ko J. Roles of CD133 in microvesicle formation and oncoprotein trafficking in colon cancer. FASEB J. 2019;33:4248–60.

    Article  CAS  PubMed  Google Scholar 

  51. Haldavnekar R, Venkatakrishnan K, Tan B. Cancer Stem Cell Derived Extracellular vesicles with self-functionalized 3D Nanosensor for Real-Time Cancer diagnosis: eliminating the roadblocks in Liquid Biopsy. ACS Nano. 2022;16:12226–43.

    Article  CAS  PubMed  Google Scholar 

  52. Refaat A, Yap ML, Pietersz G, Walsh APG, Zeller J, Del Rosal B, Wang X, Peter K. In vivo fluorescence imaging: success in preclinical imaging paves the way for clinical applications. J Nanobiotechnol. 2022;20:450.

    Article  Google Scholar 

  53. Gaedicke S, Braun F, Prasad S, Machein M, Firat E, Hettich M, Gudihal R, Zhu X, Klingner K, Schuler J, et al. Noninvasive positron emission tomography and fluorescence imaging of CD133 + tumor stem cells. Proc Natl Acad Sci U S A. 2014;111:E692–701.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Hu K, Ma X, Xie L, Zhang Y, Hanyu M, Obata H, Zhang L, Nagatsu K, Suzuki H, Shi R, et al. Development of a stable peptide-based PET Tracer for detecting CD133-Expressing Cancer cells. ACS Omega. 2022;7:334–41.

    Article  CAS  PubMed  Google Scholar 

  55. Chen YW, Liou GG, Pan HB, Tseng HH, Hung YT, Chou CP. Specific detection of CD133-positive tumor cells with iron oxide nanoparticles labeling using noninvasive molecular magnetic resonance imaging. Int J Nanomed. 2015;10:6997–7018.

    CAS  Google Scholar 

  56. Kunihiro AG, Sarrett SM, Lastwika KJ, Solan JL, Pisarenko T, Keinanen O, Rodriguez C, Taverne LR, Fitzpatrick AL, Li CI, et al. CD133 as a biomarker for an autoantibody-to-ImmunoPET paradigm for the early detection of small cell Lung Cancer. J Nucl Med. 2022;63:1701–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Jung KH, Lee JH, Kim M, Lee EJ, Cho YS, Lee KH. Celecoxib-Induced modulation of Colon cancer CD133 expression occurs through AKT Inhibition and is monitored by (89)zr Immuno-PET. Mol Imaging. 2022;2022:4906934.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Weigand S, Herting F, Maisel D, Nopora A, Voss E, Schaab C, Klammer M, Tebbe A. Global quantitative phosphoproteome analysis of human tumor xenografts treated with a CD44 antagonist. Cancer Res. 2012;72:4329–39.

    Article  CAS  PubMed  Google Scholar 

  59. Jauw YWS, Huisman MC, Nayak TK, Vugts DJ, Christen R, Naegelen VM, Ruettinger D, Heil F, Lammertsma AA, Verheul HMW, et al. Assessment of target-mediated uptake with immuno-PET: analysis of a phase I clinical trial with an anti-CD44 antibody. EJNMMI Res. 2018;8:6.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Diebolder P, Mpoy C, Scott J, Huynh TT, Fields R, Spitzer D, Bandara N, Rogers BE. Preclinical evaluation of an Engineered single-chain fragment variable-fragment Crystallizable Targeting Human CD44. J Nucl Med. 2021;62:137–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Haylock AK, Spiegelberg D, Mortensen AC, Selvaraju RK, Nilvebrant J, Eriksson O, Tolmachev V, Nestor MV. Evaluation of a novel type of imaging probe based on a recombinant bivalent mini-antibody construct for detection of CD44v6-expressing squamous cell carcinoma. Int J Oncol. 2016;48:461–70.

    Article  CAS  PubMed  Google Scholar 

  62. Ishimoto T, Nagano O, Yae T, Tamada M, Motohara T, Oshima H, Oshima M, Ikeda T, Asaba R, Yagi H, et al. CD44 variant regulates redox status in cancer cells by stabilizing the xCT subunit of system xc(-) and thereby promotes tumor growth. Cancer Cell. 2011;19:387–400.

    Article  CAS  PubMed  Google Scholar 

  63. Baek S, Choi CM, Ahn SH, Lee JW, Gong G, Ryu JS, Oh SJ, Bacher-Stier C, Fels L, Koglin N, et al. Exploratory clinical trial of (4S)-4-(3-[18F]fluoropropyl)-L-glutamate for imaging xC- transporter using positron emission tomography in patients with non-small cell lung or breast cancer. Clin Cancer Res. 2012;18:5427–37.

    Article  CAS  PubMed  Google Scholar 

  64. Baek S, Mueller A, Lim YS, Lee HC, Lee YJ, Gong G, Kim JS, Ryu JS, Oh SJ, Lee SJ, et al. (4S)-4-(3-18F-fluoropropyl)-L-glutamate for imaging of xC transporter activity in hepatocellular carcinoma using PET: preclinical and exploratory clinical studies. J Nucl Med. 2013;54:117–23.

    Article  CAS  PubMed  Google Scholar 

  65. Park SY, Na SJ, Kumar M, Mosci C, Wardak M, Koglin N, Bullich S, Mueller A, Berndt M, Stephens AW, et al. Clinical evaluation of (4S)-4-(3-[(18)F]Fluoropropyl)-L-glutamate ((18)F-FSPG) for PET/CT Imaging in patients with newly diagnosed and recurrent prostate Cancer. Clin Cancer Res. 2020;26:5380–7.

    Article  CAS  PubMed  Google Scholar 

  66. Jacobson O, Weiss ID, Szajek L, Farber JM, Kiesewetter DO. 64Cu-AMD3100–a novel imaging agent for targeting chemokine receptor CXCR4. Bioorg Med Chem. 2009;17:1486–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Jacobson O, Weiss ID, Kiesewetter DO, Farber JM, Chen X. PET of tumor CXCR4 expression with 4-18F-T140. J Nucl Med. 2010;51:1796–804.

    Article  CAS  PubMed  Google Scholar 

  68. De Silva RA, Peyre K, Pullambhatla M, Fox JJ, Pomper MG, Nimmagadda S. Imaging CXCR4 expression in human cancer xenografts: evaluation of monocyclam 64Cu-AMD3465. J Nucl Med. 2011;52:986–93.

    Article  PubMed  Google Scholar 

  69. Gourni E, Demmer O, Schottelius M, D’Alessandria C, Schulz S, Dijkgraaf I, Schumacher U, Schwaiger M, Kessler H, Wester HJ. PET of CXCR4 expression by a (68)Ga-labeled highly specific targeted contrast agent. J Nucl Med. 2011;52:1803–10.

    Article  CAS  PubMed  Google Scholar 

  70. Hennrich U, Seyler L, Schafer M, Bauder-Wust U, Eisenhut M, Semmler W, Bauerle T. Synthesis and in vitro evaluation of 68Ga-DOTA-4-FBn-TN14003, a novel tracer for the imaging of CXCR4 expression. Bioorg Med Chem. 2012;20:1502–10.

    Article  CAS  PubMed  Google Scholar 

  71. Zhang XX, Sun Z, Guo J, Wang Z, Wu C, Niu G, Ma Y, Kiesewetter DO, Chen X. Comparison of (18)F-labeled CXCR4 antagonist peptides for PET imaging of CXCR4 expression. Mol Imaging Biol. 2013;15:758–67.

    Article  PubMed  Google Scholar 

  72. Hartimath SV, van Waarde A, Dierckx RA, de Vries EF. Evaluation of N-[(11)C]methyl-AMD3465 as a PET tracer for imaging of CXCR4 receptor expression in a C6 glioma tumor model. Mol Pharm. 2014;11:3810–7.

    Article  CAS  PubMed  Google Scholar 

  73. Wester HJ, Keller U, Schottelius M, Beer A, Philipp-Abbrederis K, Hoffmann F, Simecek J, Gerngross C, Lassmann M, Herrmann K, et al. Disclosing the CXCR4 expression in lymphoproliferative diseases by targeted molecular imaging. Theranostics. 2015;5:618–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Herrmann K, Lapa C, Wester HJ, Schottelius M, Schiepers C, Eberlein U, Bluemel C, Keller U, Knop S, Kropf S, et al. Biodistribution and radiation dosimetry for the chemokine receptor CXCR4-targeting probe 68Ga-pentixafor. J Nucl Med. 2015;56:410–6.

    Article  CAS  PubMed  Google Scholar 

  75. Philipp-Abbrederis K, Herrmann K, Knop S, Schottelius M, Eiber M, Luckerath K, Pietschmann E, Habringer S, Gerngross C, Franke K, et al. In vivo molecular imaging of chemokine receptor CXCR4 expression in patients with advanced multiple myeloma. EMBO Mol Med. 2015;7:477–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Wang Z, Zhang M, Wang L, Wang S, Kang F, Li G, Jacobson O, Niu G, Yang W, Wang J, Chen X. Prospective study of (68)Ga-NOTA-NFB: Radiation Dosimetry in healthy volunteers and first application in Glioma patients. Theranostics. 2015;5:882–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Yan X, Niu G, Wang Z, Yang X, Kiesewetter DO, Jacobson O, Shen B, Chen X. Al[18F]NOTA-T140 peptide for Noninvasive visualization of CXCR4 expression. Mol Imaging Biol. 2016;18:135–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Lapa C, Schreder M, Schirbel A, Samnick S, Kortum KM, Herrmann K, Kropf S, Einsele H, Buck AK, Wester HJ, et al. [(68)Ga]Pentixafor-PET/CT for imaging of chemokine receptor CXCR4 expression in multiple myeloma - comparison to [(18)F]FDG and laboratory values. Theranostics. 2017;7:205–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Pan Q, Cao X, Luo Y, Li J, Feng J, Li F. Chemokine receptor-4 targeted PET/CT with (68)Ga-Pentixafor in assessment of newly diagnosed multiple myeloma: comparison to (18)F-FDG PET/CT. Eur J Nucl Med Mol Imaging. 2020;47:537–46.

    Article  CAS  PubMed  Google Scholar 

  80. Shekhawat AS, Singh B, Malhotra P, Watts A, Basher R, Kaur H, Hooda M, Radotra BD. Imaging CXCR4 receptors expression for staging multiple myeloma by using (68)Ga-Pentixafor PET/CT: comparison with (18)F-FDG PET/CT. Br J Radiol. 2022;95:20211272.

    Article  PubMed  PubMed Central  Google Scholar 

  81. Mayerhoefer ME, Raderer M, Lamm W, Pichler V, Pfaff S, Weber M, Kiesewetter B, Hacker M, Kazianka L, Staber PB, et al. CXCR4 PET imaging of mantle cell lymphoma using [(68)Ga]Pentixafor: comparison with [(18)F]FDG-PET. Theranostics. 2021;11:567–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Mayerhoefer ME, Raderer M, Weber M, Lamm W, Kiesewetter B, Hacker M, Nics L, Schmitl S, Leithner D, Wester HJ, Haug A. 68Ga-Pentixafor PET/MRI for Treatment Response Assessment in Mantle Cell Lymphoma: comparison between changes in Lesion CXCR4 expression on PET and lesion size and diffusivity on MRI. Clin Nucl Med. 2023;48:557–62.

    Article  PubMed  Google Scholar 

  83. Duell J, Krummenast F, Schirbel A, Klassen P, Samnick S, Rauert-Wunderlich H, Rasche L, Buck AK, Wester HJ, Rosenwald A, et al. Improved primary staging of Marginal-Zone lymphoma by Addition of CXCR4-Directed PET/CT. J Nucl Med. 2021;62:1415–21.

    Article  CAS  PubMed  Google Scholar 

  84. Herhaus P, Lipkova J, Lammer F, Yakushev I, Vag T, Slotta-Huspenina J, Habringer S, Lapa C, Pukrop T, Hellwig D, et al. CXCR4-Targeted PET imaging of Central Nervous System B-Cell Lymphoma. J Nucl Med. 2020;61:1765–71.

    Article  CAS  PubMed  Google Scholar 

  85. Starzer AM, Berghoff AS, Traub-Weidinger T, Haug AR, Widhalm G, Hacker M, Rausch I, Preusser M, Mayerhoefer ME. Assessment of Central Nervous System Lymphoma based on CXCR4 expression in vivo using 68Ga-Pentixafor PET/MRI. Clin Nucl Med. 2021;46:16–20.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Chen Z, Yang A, Zhang J, Chen A, Zhang Y, Huang C, Chen S, Yao S, Miao W. CXCR4-Directed PET/CT with [(68)Ga]Pentixafor in Central Nervous System Lymphoma: a comparison with [(18)F]FDG PET/CT. Mol Imaging Biol. 2022;24:416–24.

    Article  CAS  PubMed  Google Scholar 

  87. Luo Y, Cao X, Pan Q, Li J, Feng J, Li F. (68)Ga-Pentixafor PET/CT for Imaging of Chemokine Receptor 4 expression in Waldenstrom Macroglobulinemia/Lymphoplasmacytic Lymphoma: comparison to (18)F-FDG PET/CT. J Nucl Med. 2019;60:1724–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Pan Q, Cao X, Luo Y, Li J, Li F. Semi-quantitative measurements of chemokine receptor 4-targeted (68)Ga-pentixafor PET/CT in response assessment of Waldenstrom macroglobulinemia/lymphoplasmacytic lymphoma. EJNMMI Res. 2021;11:110.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Haug AR, Leisser A, Wadsak W, Mitterhauser M, Pfaff S, Kropf S, Wester HJ, Hacker M, Hartenbach M, Kiesewetter-Wiederkehr B, et al. Prospective non-invasive evaluation of CXCR4 expression for the diagnosis of MALT lymphoma using [(68)Ga]Ga-Pentixafor-PET/MRI. Theranostics. 2019;9:3653–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Mayerhoefer ME, Raderer M, Lamm W, Weber M, Kiesewetter B, Rohrbeck J, Simonitsch-Klupp I, Hacker M, Leisser A, Nics L, et al. CXCR4 PET/MRI for follow-up of gastric mucosa-associated lymphoid tissue lymphoma after first-line Helicobacter pylori eradication. Blood. 2022;139:240–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Lapa C, Luckerath K, Kleinlein I, Monoranu CM, Linsenmann T, Kessler AF, Rudelius M, Kropf S, Buck AK, Ernestus RI, et al. (68)Ga-Pentixafor-PET/CT for Imaging of Chemokine Receptor 4 expression in Glioblastoma. Theranostics. 2016;6:428–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Jacobs SM, Wesseling P, de Keizer B, Tolboom N, Ververs FFT, Krijger GC, Westerman BA, Snijders TJ, Robe PA, van der Kolk AG. CXCR4 expression in glioblastoma tissue and the potential for PET imaging and treatment with [(68)Ga]Ga-Pentixafor /[(177)Lu]Lu-Pentixather. Eur J Nucl Med Mol Imaging. 2022;49:481–91.

    Article  CAS  PubMed  Google Scholar 

  93. Buck AK, Haug A, Dreher N, Lambertini A, Higuchi T, Lapa C, Weich A, Pomper MG, Wester HJ, Zehndner A, et al. Imaging of C-X-C motif chemokine receptor 4 expression in 690 patients with solid or hematologic neoplasms using (68)Ga-Pentixafor PET. J Nucl Med. 2022;63:1687–92.

    CAS  PubMed  Google Scholar 

  94. Vag T, Gerngross C, Herhaus P, Eiber M, Philipp-Abbrederis K, Graner FP, Ettl J, Keller U, Wester HJ, Schwaiger M. First experience with chemokine receptor CXCR4-Targeted PET imaging of patients with solid cancers. J Nucl Med. 2016;57:741–6.

    Article  CAS  PubMed  Google Scholar 

  95. Werner RA, Kircher S, Higuchi T, Kircher M, Schirbel A, Wester HJ, Buck AK, Pomper MG, Rowe SP, Lapa C. CXCR4-Directed Imaging in Solid tumors. Front Oncol. 2019;9:770.

    Article  PubMed  PubMed Central  Google Scholar 

  96. Serfling SE, Lapa C, Dreher N, Hartrampf PE, Rowe SP, Higuchi T, Schirbel A, Weich A, Hahner S, Fassnacht M, et al. Impact of Tumor Burden on normal organ distribution in patients imaged with CXCR4-Targeted [(68)Ga]Ga-PentixaFor PET/CT. Mol Imaging Biol. 2022;24:659–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Hartrampf PE, Kosmala A, Serfling SE, Bundschuh L, Higuchi T, Lapa C, Rowe SP, Matsusaka Y, Weich A, Buck AK, et al. Interobserver Agreement Rates on C-X-C Motif Chemokine receptor 4-Directed Molecular Imaging and Therapy. Clin Nucl Med. 2023;48:483–8.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Vag T, Steiger K, Rossmann A, Keller U, Noske A, Herhaus P, Ettl J, Niemeyer M, Wester HJ, Schwaiger M. PET imaging of chemokine receptor CXCR4 in patients with primary and recurrent breast carcinoma. EJNMMI Res. 2018;8:90.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Weich A, Werner RA, Buck AK, Hartrampf PE, Serfling SE, Scheurlen M, Wester HJ, Meining A, Kircher S, Higuchi T et al. CXCR4-Directed PET/CT in patients with newly diagnosed neuroendocrine carcinomas. Diagnostics (Basel) 2021, 11.

  100. Terris B, Cavard C, Perret C. EpCAM, a new marker for cancer stem cells in hepatocellular carcinoma. J Hepatol. 2010;52:280–1.

    Article  CAS  PubMed  Google Scholar 

  101. Eder M, Knackmuss S, Le Gall F, Reusch U, Rybin V, Little M, Haberkorn U, Mier W, Eisenhut M. 68Ga-labelled recombinant antibody variants for immuno-PET imaging of solid tumours. Eur J Nucl Med Mol Imaging. 2010;37:1397–407.

    Article  CAS  PubMed  Google Scholar 

  102. Li F, Zeng Z, Hamilton D, Zu Y, Li Z. EpCAM-Targeting Aptamer Radiotracer for Tumor-Specific PET Imaging. Bioconjug Chem. 2021;32:1139–45.

    Article  CAS  PubMed  Google Scholar 

  103. Deyev SM, Vorobyeva A, Schulga A, Abouzayed A, Gunther T, Garousi J, Konovalova E, Ding H, Graslund T, Orlova A, Tolmachev V. Effect of a radiolabel biochemical nature on tumor-targeting properties of EpCAM-binding engineered scaffold protein DARPin Ec1. Int J Biol Macromol. 2020;145:216–25.

    Article  CAS  PubMed  Google Scholar 

  104. Vorobyeva A, Konovalova E, Xu T, Schulga A, Altai M, Garousi J, Rinne SS, Orlova A, Tolmachev V, Deyev S. Feasibility of Imaging EpCAM expression in Ovarian Cancer using Radiolabeled DARPin Ec1. Int J Mol Sci 2020, 21.

  105. Vorobyeva A, Bezverkhniaia E, Konovalova E, Schulga A, Garousi J, Vorontsova O, Abouzayed A, Orlova A, Deyev S, Tolmachev V. Radionuclide Molecular Imaging of EpCAM Expression in Triple-Negative Breast Cancer Using the Scaffold Protein DARPin Ec1. Molecules 2020, 25.

  106. Singh N, Shi S, Goel S. Ultrasmall silica nanoparticles in translational biomedical research: overview and outlook. Adv Drug Deliv Rev. 2023;192:114638.

    Article  CAS  PubMed  Google Scholar 

  107. Liang S, Yao J, Liu D, Rao L, Chen X, Wang Z. Harnessing nanomaterials for Cancer Sonodynamic Immunotherapy. Adv Mater. 2023;35:e2211130.

    Article  PubMed  Google Scholar 

  108. Pan Y, Liu L, Rao L, Chen X. Nanomaterial-mediated ablation therapy for cancer stem cells. Matter. 2022;5:1367–90.

    Article  CAS  Google Scholar 

  109. Ning ST, Lee SY, Wei MF, Peng CL, Lin SY, Tsai MH, Lee PC, Shih YH, Lin CY, Luo TY, Shieh MJ. Targeting Colorectal Cancer Stem-Like cells with Anti-CD133 antibody-conjugated SN-38 nanoparticles. ACS Appl Mater Interfaces. 2016;8:17793–804.

    Article  CAS  PubMed  Google Scholar 

  110. Li Y, Shi S, Ming Y, Wang L, Li C, Luo M, Li Z, Li B, Chen J. Specific cancer stem cell-therapy by albumin nanoparticles functionalized with CD44-mediated targeting. J Nanobiotechnol. 2018;16:99.

    Article  CAS  Google Scholar 

  111. Wang Q, Zhong Y, Liu W, Wang Z, Gu L, Li X, Zheng J, Du H, Zhong Z, Xie F. Enhanced chemotherapeutic efficacy of the low-dose doxorubicin in breast cancer via nanoparticle delivery system crosslinked hyaluronic acid. Drug Deliv. 2019;26:12–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Geng Y, Amante JJ, Goel HL, Zhang X, Walker MR, Luther DC, Mercurio AM, Rotello VM. Differentiation of Cancer Stem cells through nanoparticle Surface Engineering. ACS Nano. 2020;14:15276–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Wu K, Zhang W, Chen H, Wu J, Wang X, Yang X, Liang XJ, Zhang J, Liu D. An iron oxyhydroxide-based nanosystem sensitizes ferroptosis by a three-pronged strategy in breast cancer stem cells. Acta Biomater. 2023;160:281–96.

    Article  CAS  PubMed  Google Scholar 

  114. Bostad M, Olsen CE, Peng Q, Berg K, Hogset A, Selbo PK. Light-controlled endosomal escape of the novel CD133-targeting immunotoxin AC133-saporin by photochemical internalization - A minimally invasive cancer stem cell-targeting strategy. J Control Release. 2015;206:37–48.

    Article  CAS  PubMed  Google Scholar 

  115. Lee J, Davaa E, Jiang Y, Shin KJ, Kim MH, An H, Kim J, Cho SK, Yang SG. Pheophorbide A and SN38 conjugated hyaluronan nanoparticles for photodynamic- and cascadic chemotherapy of cancer stem-like ovarian cancer. Carbohydr Polym. 2022;289:119455.

    Article  CAS  PubMed  Google Scholar 

  116. Jung HS, Koo S, Won M, An S, Park H, Sessler JL, Han J, Kim JS. Cu(ii)-BODIPY photosensitizer for CAIX overexpressed cancer stem cell therapy. Chem Sci. 2023;14:1808–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Shang Q, Zhou S, Zhou Z, Jiang Y, Luan Y. Dual cancer stem cell manipulation to enhance phototherapy against tumor progression and metastasis. J Control Release. 2021;340:282–91.

    Article  CAS  PubMed  Google Scholar 

  118. Wang J, Yang B, Lv C, Chen T, Sun L, Sun L, Hao J, Ding F, Wang T, Jiang J, Qin Y. Amino porphyrin-peptide assemblies induce ribosome damage and cancer stem cell inhibition for an enhanced photodynamic therapy. Biomaterials. 2022;289:121812.

    Article  CAS  PubMed  Google Scholar 

  119. Dong X, Yin W, Zhang X, Zhu S, He X, Yu J, Xie J, Guo Z, Yan L, Liu X, et al. Intelligent MoS(2) nanotheranostic for targeted and Enzyme-/pH-/NIR-Responsive drug delivery to overcome Cancer Chemotherapy Resistance guided by PET imaging. ACS Appl Mater Interfaces. 2018;10:4271–84.

    Article  CAS  PubMed  Google Scholar 

  120. Liu L, Pan Y, Zhao C, Huang P, Chen X, Rao L. Boosting checkpoint immunotherapy with Biomaterials. ACS Nano. 2023;17:3225–58.

    Article  CAS  PubMed  Google Scholar 

  121. Liu Q, Sun Z, Duo Y, Yin L, Lv K, Yue L, Meng Q-F, Nie D, Chen J, Luo D, et al. Chromium nanoparticles improve macrophage and T cell infiltration for Cancer Immunotherapy. ACS Mater Lett. 2023;5:1738–51.

    Article  CAS  Google Scholar 

  122. Lang T, Liu Y, Zheng Z, Ran W, Zhai Y, Yin Q, Zhang P, Li Y. Cocktail Strategy based on Spatio-temporally controlled Nano device improves therapy of breast Cancer. Adv Mater. 2019;31:e1806202.

    Article  PubMed  Google Scholar 

  123. Kim S, Heo R, Song SH, Song KH, Shin JM, Oh SJ, Lee HJ, Chung JE, Park JH, Kim TW. PD-L1 siRNA-hyaluronic acid conjugate for dual-targeted cancer immunotherapy. J Control Release. 2022;346:226–39.

    Article  CAS  PubMed  Google Scholar 

  124. Cheng Y, Wang C, Wang H, Zhang Z, Yang X, Dong Y, Ma L, Luo J. Combination of an autophagy inhibitor with immunoadjuvants and an anti-PD-L1 antibody in multifunctional nanoparticles for enhanced breast cancer immunotherapy. BMC Med. 2022;20:411.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Hassani Najafabadi A, Zhang J, Aikins ME, Najaf Abadi ZI, Liao F, Qin Y, Okeke EB, Scheetz LM, Nam J, Xu Y, et al. Cancer Immunotherapy via Targeting Cancer Stem cells using vaccine nanodiscs. Nano Lett. 2020;20:7783–92.

    Article  CAS  PubMed  Google Scholar 

  126. Aikins ME, Qin Y, Dobson HE, Najafabadi AH, Lyu K, Xu Y, Xin Y, Schwendeman A, Wicha MS, Chang AE, et al. Cancer stem cell antigen nanodisc cocktail elicits anti-tumor immune responses in melanoma. J Control Release. 2022;351:872–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Chen C, Jing W, Chen Y, Wang G, Abdalla M, Gao L, Han M, Shi C, Li A, Sun P, et al. Intracavity generation of glioma stem cell-specific CAR macrophages primes locoregional immunity for postoperative glioblastoma therapy. Sci Transl Med. 2022;14:eabn1128.

    Article  CAS  PubMed  Google Scholar 

  128. Weng D, Jin X, Qin S, Lan X, Chen C, Sun X, She X, Dong C, An R. Radioimmunotherapy for CD133(+) colonic cancer stem cells inhibits tumor development in nude mice. Oncotarget. 2017;8:44004–14.

    Article  PubMed  PubMed Central  Google Scholar 

  129. She X, Qin S, Jing B, Jin X, Sun X, Lan X, An R. Radiotheranostic Targeting Cancer Stem cells in human colorectal Cancer xenografts. Mol Imaging Biol. 2020;22:1043–53.

    Article  CAS  PubMed  Google Scholar 

  130. Sternberg CN, Magill GB, Cheng EW, Hollander P. Phase II trial of menogarol in the treatment of advanced adenocarcinoma of the pancreas. Am J Clin Oncol. 1988;11:174–6.

    Article  CAS  PubMed  Google Scholar 

  131. Herrmann K, Schottelius M, Lapa C, Osl T, Poschenrieder A, Hanscheid H, Luckerath K, Schreder M, Bluemel C, Knott M, et al. First-in-human experience of CXCR4-Directed endoradiotherapy with 177Lu- and 90Y-Labeled Pentixather in Advanced-Stage multiple myeloma with extensive intra- and Extramedullary Disease. J Nucl Med. 2016;57:248–51.

    Article  CAS  PubMed  Google Scholar 

  132. Schottelius M, Osl T, Poschenrieder A, Hoffmann F, Beykan S, Hanscheid H, Schirbel A, Buck AK, Kropf S, Schwaiger M, et al. [(177)Lu]pentixather: Comprehensive Preclinical characterization of a first CXCR4-directed Endoradiotherapeutic Agent. Theranostics. 2017;7:2350–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Lapa C, Hanscheid H, Kircher M, Schirbel A, Wunderlich G, Werner RA, Samnick S, Kotzerke J, Einsele H, Buck AK, et al. Feasibility of CXCR4-Directed Radioligand therapy in advanced diffuse large B-Cell lymphoma. J Nucl Med. 2019;60:60–4.

    Article  CAS  PubMed  Google Scholar 

  134. Habringer S, Lapa C, Herhaus P, Schottelius M, Istvanffy R, Steiger K, Slotta-Huspenina J, Schirbel A, Hanscheid H, Kircher S, et al. Dual targeting of Acute Leukemia and supporting Niche by CXCR4-Directed Theranostics. Theranostics. 2018;8:369–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Buck AK, Grigoleit GU, Kraus S, Schirbel A, Heinsch M, Dreher N, Higuchi T, Lapa C, Hanscheid H, Samnick S, et al. C-X-C motif chemokine receptor 4-Targeted Radioligand Therapy in patients with Advanced T-Cell Lymphoma. J Nucl Med. 2023;64:34–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Maurer S, Herhaus P, Lippenmeyer R, Hanscheid H, Kircher M, Schirbel A, Maurer HC, Buck AK, Wester HJ, Einsele H, et al. Side effects of CXC-Chemokine receptor 4-Directed Endoradiotherapy with Pentixather before hematopoietic stem cell transplantation. J Nucl Med. 2019;60:1399–405.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Osl T, Schmidt A, Schwaiger M, Schottelius M, Wester HJ. A new class of PentixaFor- and PentixaTher-based theranostic agents with enhanced CXCR4-targeting efficiency. Theranostics. 2020;10:8264–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by National Natural Science Foundation of China (82002766, 81702570, 82102121), National Key Research and Development Program of China (2022YFB3808200), Key Research and Development Program of Hubei Province (2021BCA118), National Health Commission Central Asia High Incidence Disease Prevention and Control Key Laboratory Open Project (KF202202).

Author information

Authors and Affiliations

Authors

Contributions

SS and QY contributed equally to this work. YZ and DJ proposed the project; SS and QY prepared and revised the manuscript. All authors have reviewed and approved the final version of the manuscript.

Corresponding authors

Correspondence to Dawei Jiang or Yuan Zhang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

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

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sun, S., Yang, Q., Jiang, D. et al. Nanobiotechnology augmented cancer stem cell guided management of cancer: liquid-biopsy, imaging, and treatment. J Nanobiotechnol 22, 176 (2024). https://doi.org/10.1186/s12951-024-02432-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12951-024-02432-5

Keywords