ArticlePublisher preview available

Bifunctional Therapeutic Peptide Assembled Nanoparticles Exerting Improved Activities of Tumor Vessel Normalization and Immune Checkpoint Inhibition

Wiley
Advanced Healthcare Materials
Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract and Figures

The effectiveness of cancer immunotherapy is impaired by the dysfunctional vasculature of tumors. Created hypoxia zones and limited delivery of cytotoxic immune cells help to have immune resistance in tumor tissue. Structural and functional normalization of abnormal tumor vasculature provide vessels for more perfusion efficiency and drug delivery that result in alleviating the hypoxia in the tumor site and increasing infiltration of antitumor T cells. Taking advantage of peptide amphiphiles, herein, a novel peptide amphiphile nanoparticle composed of an antiangiogenic peptide (FSEC) and an immune checkpoint blocking peptide (DPPA) is designed and characterized. FSEC peptide is known to be involved in vessel normalization of tumors in vivo. DPPA is an inhibitory peptide of the PD‐1/PD‐L1 immune checkpoint pathway. The peptide amphiphile nanoparticle sets out to test whether simultaneous modulation of tumor vasculature and immune systems in the tumor microenvironment has a synergistic effect on tumor suppression. Increased intratumoral infiltration of immune cells following vascular normalization, and simultaneously blocking the immune checkpoint function of PD‐L1 reactivates effective immune responses to the tumors. In summary, the current study provides a new perspective on the regulation of tumor vessel normalization and immunotherapy based on functional peptide nanoparticles as nanomedicine for improved therapeutic purposes.
In vivo evaluation of designed sequences effect on the normalization of tumor vessel structures, leakiness, and hypoxia. a) FSEC peptide sequence act as a specific VEGF receptor antagonist and promotes vessel normalization through inhibition of angiogenesis. Double immunofluorescence staining of pericytes (anti‐αSMA antibody, green) and endothelial cells (anti‐CD31 antibody, red) visualized the regained vascular organizations in groups that have been treated by FSEC peptide sequence and in particular by FD nanoparticles. b) Vascular maturity was calculated through the ratio of pericytes coverage to endothelial cells. c) Changes in vascular leakage were evaluated by FITC–dextran in vivo 14 days after treatment with peptide sequences. Scale bars = 100 µm. d) Relative vessel leakiness. e) The modified Miles assay was used to assess vascular permeability in the tumor. Evans blue dye concentration shows significantly decreased vessel leakiness after the treatment of mice using FD nanoparticles. f) Confocal microscopy images of vessels perfused with FITC–lectin. Perfusion of vessels after normalization was increased meaningfully in FD nanoparticle treated group. Treatment with other sequences cannot normalize vessels, which represent dilation and leaked structure similar to untreated tumors. Scale bar = 100 µm. g) Relative vessel perfusion intensity. h) Immunohistochemical staining for HIF1α in tumor tissues of treatment groups. Hypoxic areas significantly decreased in groups treated by the FSEC peptide sequence or FD nanoparticles compared to other control groups. i) Decreased expression levels of HIF1α in the tumors after treatment by FSEC peptide sequences and FD nanoparticles indicate better tissue oxygenation and less hypoxic effect as a consequence of vessel normalization. Scale bar = 50 µm. The experiments were repeated three times.
… 
This content is subject to copyright. Terms and conditions apply.
RESEARCH ARTICLE
www.advhealthmat.de
Bifunctional Therapeutic Peptide Assembled Nanoparticles
Exerting Improved Activities of Tumor Vessel Normalization
and Immune Checkpoint Inhibition
Mohammad Taleb, Mona Atabakhshi-Kashi, Yazhou Wang, Hamideh Rezvani Alanagh,
Zeinab Farhadi Sabet, Fenfen Li, Keman Cheng, Chen Li, Yingqiu Qi, Guangjun Nie,*
and Zhao Ying*
The effectiveness of cancer immunotherapy is impaired by the dysfunctional
vasculature of tumors. Created hypoxia zones and limited delivery of cytotoxic
immune cells help to have immune resistance in tumor tissue. Structural and
functional normalization of abnormal tumor vasculature provide vessels for
more perfusion efficiency and drug delivery that result in alleviating the
hypoxia in the tumor site and increasing infiltration of antitumor T cells.
Taking advantage of peptide amphiphiles, herein, a novel peptide amphiphile
nanoparticle composed of an antiangiogenic peptide (FSEC) and an immune
checkpoint blocking peptide (DPPA) is designed and characterized. FSEC
peptide is known to be involved in vessel normalization of tumors in vivo.
DPPA is an inhibitory peptide of the PD-1/PD-L1 immune checkpoint pathway.
The peptide amphiphile nanoparticle sets out to test whether simultaneous
modulation of tumor vasculature and immune systems in the tumor
microenvironment has a synergistic effect on tumor suppression. Increased
intratumoral infiltration of immune cells following vascular normalization,
and simultaneously blocking the immune checkpoint function of PD-L1
reactivates effective immune responses to the tumors. In summary, the
current study provides a new perspective on the regulation of tumor vessel
normalization and immunotherapy based on functional peptide nanoparticles
as nanomedicine for improved therapeutic purposes.
1. Introduction
Immune-oncology therapies have emerged as an eective treat-
ment in cancer patients, engaging the immune system of
M. Taleb, Dr. M. Atabakhshi-Kashi, Dr. Y. Wang, Dr. H. Rezvani Alanagh,
Z.FarhadiSabet,F.Li,Dr.K.Cheng,C.Li,Prof.G.Nie,Prof.Z.Ying
CAS Key Laboratory for Biomedical Eects of Nanomaterials and
Nanosafety
CAS Center of Excellence in Nanoscience
National Center for Nanoscience and Technology
Beijing , P. R. China
E-mail: niegj@nanoctr.cn; zhaoying@nanoctr.cn
The ORCID identification number(s) for the author(s) of this article
can be found under https://doi.org/./adhm.
DOI: 10.1002/adhm.202100051
themselves to recognize and eradicate tu-
mor cells.[] In this regard, targeting in-
hibitory receptors on cytotoxic T cells
and/or tumor cells can constructively rein-
vigorate antitumor immune responses in
the tumor microenvironment (TME) and
partially modulate the immunosuppres-
sive microenvironment of tumors.[, ] Pro-
grammed cell death ligand- (PD-L) is
known as one of these inhibitory recep-
tors on cancerous cells which engages in
the PD-/PD-L axis and subsequently pre-
vents the killing capacity of T cells.[] In-
terference with the PD-/PD-L interac-
tion using immune checkpoint inhibitors
(ICIs) such as monoclonal antibodies or
specific peptide sequences have an eec-
tive impact on the enhancement of antitu-
mor immune responses. Rather than con-
ventional treatment methods in particular
cytotoxic chemotherapy and radiation ther-
apy, immune-mediated tumor therapy by
ICIs increase survival advantage and pro-
vide more durable clinical outcomes.[, ]
However, most of the patients do not drive
long-term benefits from ICIs monother-
apy because of acquired resistance that is
established by tumor-associated factors.[]
According to the clinical trials, inadequate intratumoral infiltra-
tion of immune cells and the diminished oxygen availability (hy-
poxia) in TME that raised from abnormal tumor vasculatures are
concomitant with ineective responses to ICIs.[, ]
M. Taleb, Dr. H. Rezvani Alanagh, Z. Farhadi Sabet, C. Li, Prof. G. Nie,
Prof. Z. Ying
Center of Materials Science and Optoelectronics Engineering
University of Chinese Academy of Sciences
Beijing , P. R. China
Dr.Y.Qi
School of Basic Medical Science
Zhengzhou University
Henan , China
Prof. G. Nie, Prof. Z. Ying
GBA Research Innovation Institute for Nanotechnology
Guangdong , P. R. China
Adv. Healthcare Mater. 2021,10,  ©  Wiley-VCH GmbH
2100051 (1 of 13)
... The fundamental reason for the low accumulation of drugs within the tumor is the insufficient blood perfusion within the tumor interior [11]. Studies have revealed that the local capillary venous pressure within tumors is approximately 20 mm Hg, while the interstitial pressure ranges from 20 to 130 mm Hg [12]. ...
Article
Full-text available
Background Elevated interstitial fluid pressure within tumors, resulting from impaired lymphatic drainage, constitutes a critical barrier to effective drug penetration and therapeutic outcomes. Results In this study, based on the photosynthetic characteristics of algae, an active drug carrier (CP@ICG) derived from Chlorella pyrenoidosa (CP) was designed and constructed. Leveraging the hypoxia tropism and phototropism exhibited by CP, we achieved targeted transport of the carrier to tumor sites. Additionally, dual near-infrared (NIR) irradiation at the tumor site facilitated photosynthesis in CP, enabling the breakdown of excessive intratumoral interstitial fluid by generating oxygen from water decomposition. This process effectively reduced the interstitial pressure, thereby promoting enhanced perfusion of blood into the tumor, significantly improving deep-seated penetration of chemotherapeutic agents, and alleviating tumor hypoxia. Conclusions CP@ICG demonstrated a combined effect of photothermal/photodynamic/starvation therapy, exhibiting excellent in vitro/in vivo anti-tumor efficacy and favorable biocompatibility. This work provides a scientific foundation for the application of microbial-enhanced intratumoral drug delivery and tumor therapy. Graphical Abstract
... An increase in tumour-infiltrating NK cells and CD8+ T cells was then confirmed. 128 As another example, heparin sulfatase 1-expressing bacteria conjugated with NPs carrying doxorubicin can actively colonise tumour sites and dampen cancer angiogenesis and metastasis. Mechanistically, heparin sulfatase 1 can remove the sulphate of heparan sulfate proteoglycan core proteins to disturb its regulation on vascular development. ...
Article
Full-text available
Background: The immune system plays a pivotal role in the initiation, evolution, invasion and metastasis of cancer. Therapeutics aiming at modulating or boosting anticancer immune responses have experienced immense advances during the past decades, for example, anti-PD-1/PD-L1 monoclonal antibodies. Main body: Concomitant with advancements in the understanding of novel mechanisms of action, conventional or emerging drugs bearing the potential to be repurposed for enhancing anticancer immunity have been identified. Meanwhile, ongoing advances in drug delivery systems enable us to utilise novel therapeutic strategies and impart drugs with fresh modes of action in tumour immunology. Conclusion: Herein, we systemically review these kinds of drugs and delivery systems that can unleash the anticancer response through various aspects, including immune recognition, activation, infiltration and tumour killing. We also discuss the current caveats and future directions of these emerging strategies.
Article
Full-text available
With the rapid development of the fields of tumor biology and immunology, tumor immunotherapy has been used in clinical practice and has demonstrated significant therapeutic potential, particularly for treating tumors that do not respond to standard treatment options. Despite its advances, immunotherapy still has limitations, such as poor clinical response rates and differences in individual patient responses, largely because tumor tissues have strong immunosuppressive microenvironments. Many tumors have a tumor microenvironment (TME) that is characterized by hypoxia, low pH, and substantial numbers of immunosuppressive cells, and these are the main factors limiting the efficacy of antitumor immunotherapy. The TME is crucial to the occurrence, growth, and metastasis of tumors. Therefore, numerous studies have been devoted to improving the effects of immunotherapy by remodeling the TME. Effective regulation of the TME and reversal of immunosuppressive conditions are effective strategies for improving tumor immunotherapy. The use of multidrug combinations to improve the TME is an efficient way to enhance antitumor immune efficacy. However, the inability to effectively target drugs decreases therapeutic effects and causes toxic side effects. Nanodrug delivery carriers have the advantageous ability to enhance drug bioavailability and improve drug targeting. Importantly, they can also regulate the TME and deliver large or small therapeutic molecules to decrease the inhibitory effect of the TME on immune cells. Therefore, nanomedicine has great potential for reprogramming immunosuppressive microenvironments and represents a new immunotherapeutic strategy. Therefore, this article reviews strategies for improving the TME and summarizes research on synergistic nanomedicine approaches that enhance the efficacy of tumor immunotherapy.
Article
Full-text available
At present, tumor metastasis still remains the leading contributor to high recurrence and mortality in cancer patients. There have been no clinically effective therapeutic strategies for treating patients with metastatic cancer. In recent years, a growing body of evidence has shown that the pre-metastatic niche (PMN) plays a crucial role in driving tumor metastasis. Nevertheless, a clear and detailed understanding of the formation of PMN is still lacking given the fact that PMN formation involves in a wealth of complicated communications and underlying mechanisms between primary tumors and metastatic target organs. Despite that the roles of numerous components including tumor exosomes and extracellular vesicles in influencing the evolution of PMN have been well documented, the involvement of cancer-associated fibroblasts (CAFs) in the tumor microenvironment for controlling PMN formation is frequently overlooked. It has been increasingly recognized that fibroblasts trigger the formation of PMN by virtue of modulating exosomes, metabolism and so on. In this review, we mainly summarize the underlying mechanisms of fibroblasts from diverse origins in exerting impacts on PMN evolution, and further highlight the prospective strategies for targeting fibroblasts to prevent PMN formation.
Article
Full-text available
Immunotherapy is a promising cancer therapeutic strategy. However, the “cold” tumor immune microenvironment (TIME), characterized by insufficient immune cell infiltration and immunosuppressive status, limits the efficacy of immunotherapy. Tumor vascular abnormalities due to defective pericyte coverage are gradually recognized as a profound determinant in “cold” TIME establishment by hindering immune cell trafficking. Recently, several vascular normalization strategies by improving pericyte coverage have been reported, whereas have unsatisfactory efficacy and high rates of resistance. Herein, a combinatorial strategy to induce tumor vasculature‐targeted pericyte recruitment and zinc ion‐mediated immune activation with a platelet‐derived growth factor B (PDGFB)‐loaded, cyclo (Arg‐Gly‐Asp‐D‐Phe‐Lys)‐modified zeolitic imidazolate framework 8 (PDGFB@ZIF8‐RGD) nanoplatform is proposed. PDGFB@ZIF8‐RGD effectively induced tumor vascular normalization, which facilitated trafficking and infiltration of immune effector cells, including natural killer (NK) cells, M1‐like macrophages and CD8⁺ T cells, into tumor microenvironment. Simultaneously, vascular normalization promoted the accumulation of zinc ions inside tumors to trigger effector cell immune activation and effector molecule production. The synergy between these two effects endowed PDGFB@ZIF8‐RGD with superior capabilities in reprogramming the “cold” TIME to a “hot” TIME, thereby initiating robust antitumor immunity and suppressing tumor growth. This combinatorial strategy for improving immune effector cell infiltration and activation is a promising paradigm for solid tumor immunotherapy.
Article
Photoimmunotherapy (PIT) is an emerging therapeutic approach that integrates phototherapy and immunotherapy to eliminate primary tumors under an appropriate dosage of local light irradiation, while simultaneously preventing tumor metastasis and recurrence by activating the host antitumor immune response. Tumor-responsive dynamic nanoassemblies (TDNs) have evolved from being a mere curiosity to a promising platform for high-performance PIT. However, the dynamic nano-bio interaction between TDNs and tumor microenvironment remains poorly understood, which shall be critical for precise control of TDNs assembling/disassembling behavior and superior PIT efficacy. To deepen the understanding of the structure–function relationship of TDNs, this review introduces the rational design, nano-bio interactions, and controllable functionalities of cutting-edge TDNs for enhanced PIT. Moreover, the synergetic mechanism between TDNs-based PIT and immunomodulatory agents-mediated immunomodulation is particularly emphasized. Finally, the challenges and future perspectives in this emerging field are assessed.
Article
Full-text available
Natural, extracellular membrane vesicles secreted by Gram-negative bacteria, outer membrane vesicles (OMVs), contain numerous pathogen-associated molecular patterns which can activate systemic immune responses. Previous studies have shown that OMVs induce strong IFN-γ- and T cell-mediated anti-tumor effects in mice. However, IFN-γ is known to upregulate immunosuppressive factors in the tumor microenvironment, especially the immune checkpoint programmed death 1 ligand 1 (PD-L1), which may hamper T cell function and limit immunotherapeutic effectiveness. Here, we report the development of genetically engineered OMVs whose surface has been modified by insertion of the ectodomain of programmed death 1 (PD1). This genetic modification does not affect the ability of OMVs to trigger immune activation. More importantly, the engineered OMV-PD1 can bind to PD-L1 on the tumor cell surface and facilitate its internalization and reduction, thereby protecting T cells from the PD1/PD-L1 immune inhibitory axis. Through the combined effects of immune activation and checkpoint suppression, the engineered OMVs drive the accumulation of effector T cells in the tumor, which, in turn, leads to a greater impairment of tumor growth, compared with not only native OMVs but also the commonly used PD-L1 antibody. In conclusion, this work demonstrates the potential of bioengineered OMVs as effective immunotherapeutic agents that can comprehensively regulate the tumor immune microenvironment to effect markedly increased anti-tumor efficacy.
Article
Full-text available
Advances in immuno‐oncology have significantly improved cancer therapy outcomes. Unfortunately, potent responses to immunotherapy have only been achieved in subsets of patients. Nanomedicines and biomaterials have enormous potential for making immunotherapy work better in more patients, which has been suggested by a large number of preclinical studies. However, it remains unclear whether and how these materials will really benefit cancer patients treated by immunotherapy. Therefore, reviewing current clinical studies on nanomedicines and biomaterials in immuno‐oncology may provide valuable information for future translation research. In this work, the clinical landscape of cancer immunotherapy involving nanomedicines and biomaterials, mainly Abraxane, Doxil, messenger ribonucleic acid (mRNA) nanovaccines, and a biomaterial scaffold‐supported autologous vaccine, is reviewed. Clinical results demonstrate the high potential of nano/biomaterials in immunotherapy, achieved by rationalized utilization of their targeted delivery and reduced toxicities/side effects. Future clinical translation in this field will likely benefit from designing rational combinations of nanomedicines and biomaterials with the right immunotherapeutics applied in the right patients/tumors, and this should be achieved with the help of immune biomarker analysis. By rationally utilizing the unique features and advantages of nanomedicines and biomaterials, they will highly impact patient care in combination with immunotherapy.
Article
Full-text available
img src=” https://s3.amazonaws.com/production.scholastica/article/13444/medium/prnano_442020_background_figure.jpg?1592934354”> Precisely selective interactions of peptides with their unique binding partners represent an out-standing starting point for designing novel therapeutics. It is well established that peptides with a variety of critical physiological functions and specific mechanisms of action offer distinct ad-vantages, including excellent safety and higher efficiency over traditional small molecule thera-peutics. Certain intrinsic weaknesses of naturally occurring peptides such as negligible plasma half-life, low bioavailability, and potential immunogenicity have limited their administration as medicines. Nanotechnology has expanded several promising strategies to address the limitations associated with therapeutic peptides. This review aims to perform a state-of-the-art summary of the strategies that are actively used to develop efficient formulations of nanosystem based pep-tide medicines. We first focus on the recent advances and updates on peptide-based nanomedi-cines. Then we indicate how nanosystems improved the functionality of therapeutic peptides and what the future opportunities and challenges of developments in the field of therapeutic pep-tides are. Potential noninvasive delivery platforms for peptide incorporated nanoparticles through alternative administration routes are also discussed.
Article
Full-text available
Tumor-associated macrophages (TAMs) as immune cells within the tumor microenvironment have gained much interests as basic science regarding their roles in tumor progression unfolds. Better understanding of their polarization into pro-tumoral phenotype to promote tumor growth, tumor angiogenesis, immune evasion, and tumor metastasis has prompted various studies to investigate their clinical significance as a biomarker of predictive and prognostic value across different cancer types. Yet, the methodologies to investigate the polarization phenomena in solid tumor tissue vary. Nonetheless, quantifying the ratio of M1 to M2 TAMs has emerged to be a prevailing parameter to evaluate this polarization phenomena for clinical application. This mini-review focuses on recent studies exploring clinical significance of M1/M2 TAM ratio in human cancer tissue and critically evaluates the technicalities and challenges in quantifying this parameter for routine clinical practice. Immunohistochemistry appears to be the preferred methodology for M1/M2 TAM evaluation as it is readily available in clinical laboratories, albeit with certain limitations. Recommendations are made to standardize the quantification of TAMs for better transition into clinical practice and for better comparison among studies in various populations of patients and cancer types.
Article
Full-text available
Aim: Clinical resistance is a complex phenomenon in major human cancers involving multifactorial mechanisms, and hypoxia is one of the key components that affect the cellular expression program and lead to therapy resistance. The present study aimed to summarize the role of hypoxia in cancer therapy by regulating the tumor microenvironment (TME) and to highlight the potential of hypoxia-targeted therapy. Methods: Relevant published studies were retrieved from PubMed, Web of Science, and Embase using keywords such as hypoxia, cancer therapy, resistance, TME, cancer, apoptosis, DNA damage, autophagy, p53, and other similar terms. Results: Recent studies have shown that hypoxia is associated with poor prognosis in patients by regulating the TME. It confers resistance to conventional therapies through a number of signaling pathways in apoptosis, autophagy, DNA damage, mitochondrial activity, p53, and drug efflux. Conclusion: Hypoxia targeting might be relevant to overcome hypoxia-associated resistance in cancer treatment.
Article
Full-text available
Tumor vascularization occurs through several distinct biological processes, which not only vary between tumor type and anatomic location, but also occur simultaneously within the same cancer tissue. These processes are orchestrated by a range of secreted factors and signaling pathways and can involve participation of non-endothelial cells, such as progenitors or cancer stem cells. Anti-angiogenic therapies using either antibodies or tyrosine kinase inhibitors have been approved to treat several types of cancer. However, the benefit of treatment has so far been modest, some patients not responding at all and others acquiring resistance. It is becoming increasingly clear that blocking tumors from accessing the circulation is not an easy task to accomplish. Tumor vessel functionality and gene expression often differ vastly when comparing different cancer subtypes, and vessel phenotype can be markedly heterogeneous within a single tumor. Here, we summarize the current understanding of cellular and molecular mechanisms involved in tumor angiogenesis and discuss challenges and opportunities associated with vascular targeting.
Article
Multiple nanotherapeutics have been approved for patients with cancer, but their effects on survival have been modest and, in some examples, less than those of other approved therapies. At the same time, the clinical successes achieved with immunotherapy have revolutionized the treatment of multiple advanced-stage malignancies. However, the majority of patients do not benefit from the currently available immunotherapies and many develop immune-related adverse events. By contrast, nanomedicines can reduce — but do not eliminate — the risk of certain life-threatening toxicities. Thus, the combination of these therapeutic classes is of intense research interest. The tumour microenvironment (TME) is a major cause of the failure of both nanomedicines and immunotherapies that not only limits delivery, but also can compromise efficacy, even when agents accumulate in the TME. Coincidentally, the same TME features that impair nanomedicine delivery can also cause immunosuppression. In this Perspective, we describe TME normalization strategies that have the potential to simultaneously promote the delivery of nanomedicines and reduce immunosuppression in the TME. Then, we discuss the potential of a combined nanomedicine-based TME normalization and immunotherapeutic strategy designed to overcome each step of the cancer-immunity cycle and propose a broadly applicable ‘minimal combination’ of therapies designed to increase the number of patients with cancer who are able to benefit from immunotherapy.
Article
Significance Immunotherapy has changed the standard of care in cancer treatment, but an estimated 87% of patients currently do not derive long-term benefit from immune checkpoint blocker monotherapy. Therefore, new therapeutic strategies are needed to improve the response rates in patients who are resistant to immune checkpoint inhibition. We have developed a mathematical framework to determine how tumor microenvironment normalization strategies—specifically, vascular and stroma normalization—might improve immunotherapy efficacy. By incorporating complex interactions among various types of cancer cells, immune cells, stromal cells, and the vasculature, as well as physical mechanisms, we provide guidelines for designing effective combinatorial therapeutic strategies and point out areas for future investigation.
Article
The limited efficacy of single-agent immune checkpoint inhibitors in treating tumors has prompted investigations on their combination partners. Here, a tumor-homing indoleamine 2, 3-dioxygenase (IDO) nanoinhibitor is reported to selectively inhibit immunosuppressive IDO pathway in the tumor microenvironment. It is self-assembled from a modularly designed peptide-drug conjugate containing a hydrophilic targeting motif (arginyl-glycyl-aspartic acid; RGD), two protonatable histi-dines and an ester bond-linked hydrophobic IDO inhibitor, which exhibits pH-responsive disassembly and esterase-catalyzed drug release. Markedly, it achieved potent and persistent inhibition of intratumoral IDO activity with reduced systemic toxicity, which greatly enhanced the therapeutic efficacy of programmed cell death-ligand 1 blockade in vivo. Overall, this study provides a promising paradigm of combinatorial normalization immunotherapy by exploiting a targeted IDO nanoinhibitor to augment the anti-tumor immunity of checkpoint inhibitors.
Article
To better make nanomedicine entering the clinic, developing new rationally designed nanotherapeutics with a deeper understanding of tumor biology is required. The tumor microenvironment is similar to the inflammatory response in a healing wound, the milieu of which promotes tumor cell invasion and metastasis. Successful targeting of the microenvironmental components with effective nanotherapeutics to modulate the tumor microvessels or restore the homeostatic mechanisms in the tumor stroma will offer new hope for cancer treatment. We here highlight the progress in constructing nanotherapeutics to target or modulate the tumor microenvironment. We discuss the factors necessary for nanomedicines to become a new paradigm in cancer therapy, including the selection of drugs and therapeutic targets, controllable synthesis, and tempo‐spatial drug release.