Background
Colorectal cancer (CRC) ranks as the third most commonly diagnosed cancer and the second leading cause of cancer death worldwide. In 2022, there were over 1.926 million new CRC cases and 904,000 deaths globally, accounting for 10.0% and 9.5% of total cancer incidence and mortality, respectively. By 2040, the global CRC burden is projected to increase to 3.2 million new cases and 1.6 million deaths per year, representing a rise of 63% and 73%, respectively. In China, CRC has risen to become the second most common malignancy: approximately 590,000 new cases and 310,000 deaths were reported in 2022, with the proportion of young patients under 50 years of age increasing to 15%, significantly higher than the global average.
Although existing modalities such as chemotherapy, targeted therapy, and immunotherapy have advanced, current treatment approaches still face considerable limitations. Conventional chemotherapy (e.g., FOLFOX regimen) is associated with significant toxicity, targeted drugs (e.g., anti-EGFR monoclonal antibodies) show high resistance rates, and immunotherapy is effective in only about 5% of MSI-H patients . More critically, existing drug delivery systems commonly suffer from poor targeting, low tumor penetration, and low oral bioavailability. For example, the penetration rate of nanomedicines in solid tumors is less than 1% .
Therefore, the development of a drug delivery system that combines targeting specificity, efficient drug utilization, and low toxicity is essential to overcome current therapeutic bottlenecks in CRC and enhance the efficacy of drugs acting on difficult-to-reach targets.
Problem
While existing treatment modalities can extend the survival of colorectal cancer patients, the dose-dependent nature of therapy means that therapeutic gains are often offset by increased toxicity and enhanced drug resistance. The overall efficacy remains limited, failing to fully overcome this dilemma. Consequently, there is a pressing need for more efficient delivery methods to precisely transport drugs into cancer cells. The specific challenges are manifested in the following four aspects:
No. 1
Conventional Chemotherapy:
Non-Selective Toxicity and High Risk of Drug Resistance
Conventional chemotherapy utilizes chemical drugs to kill rapidly dividing cancer cells, but it simultaneously induces toxic side effects on normal cells. Agents like 5-FU and oxaliplatin cannot distinguish between cancerous and normal cells, leading to high incidences of bone marrow suppression and peripheral neurotoxicity. Mechanisms such as P-glycoprotein (ABCB1) efflux, TYMS amplification, or KRAS mutations further reduce intracellular drug concentrations and induce resistance. Regimens like FOLFOX/FOLFIRI require 48-hour intravenous infusion, yet the drug concentration in tumor tissue is only 10–20% of that in plasma, indicating limited local exposure . Oral administration of 5-FU is hampered by gastric acid degradation, resulting in low bioavailability.
No. 2
Targeted Drugs:
Limited Target Population and Low Delivery Efficiency
Targeted drugs specifically act on particular molecular targets in cancer cells to inhibit their growth and spread. However, their efficacy depends on specific genetic mutations, resulting in a narrow applicable patient population. Cetuximab is effective only in patients with KRAS/BRAF wild-type and left-sided primary tumors . The population eligible for trastuzumab is extremely limited, comprising only about 3–5% of CRC patients . Regarding delivery efficiency, monoclonal antibodies have a long half-life but exhibit low tissue penetration, leading to insufficient drug concentrations in deep tumor regions . Nanocarriers targeting CD44 or EGFR suffer from high off-target toxicity due to cross-expression in normal tissues and tumor microenvironment (TME) heterogeneity .
No. 3
Immunotherapy:
Narrow Indications and Systemic Toxicity
Immunotherapy works by activating the patient's own immune system to attack tumor cells. However, in metastatic colorectal cancer (mCRC), the efficacy of PD-1 inhibitors is highly dependent on the tumor's molecular subtype. Only approximately 5% of patients with dMMR/MSI-H tumors respond well to PD-1 inhibitors; for the remaining pMMR/MSS patients, the objective response rate (ORR) to PD-1 inhibitors alone is less than 5%, rendering them largely ineffective. Furthermore, following intravenous infusions every 2–3 weeks, some patients may experience varying degrees of immune-related adverse events, primarily including colitis, endocrinopathies, and other related toxicities .
No. 4
Delivery System Bottlenecks:
Barrier Limitations
The tumor microenvironment (TME) is the complex milieu surrounding tumor cells, comprising immune cells, blood vessels, and other components, which profoundly influences tumor progression. Nanomedicines are hindered by high interstitial pressure, dense collagen, and immune cell clearance within the TME, resulting in a penetration rate of merely 0.7% in solid tumors . Liposomes and exosomes face challenges such as batch-to-batch variability, difficulties in large-scale production, and high costs [12]. The colonic mucus layer, which can be up to 800 µm thick, further impedes the diffusion of orally administered nanoparticles, leading to inadequate local concentrations .
In summary, the core bottlenecks in colorectal cancer treatment are insufficient tumor targeting and high toxicity. While traditional antibodies possess targeting capability, their penetration rate in solid tumors is often below 1%; liposomes are constrained by batch variability and high cost . Therefore, there is an urgent need to develop more efficient delivery methods for precisely transporting drugs deep into tumors. Activatable Cell Penetrating Peptides (ACPPs) offer a targeted solution through an MMP-9 cleavage-de-shielding mechanism : their anionic segment inhibits transmembrane transport in normal tissues, but upon cleavage in the MMP-high TME, transmembrane transport is activated, efficiently delivering drugs into cancer cells . This ensures activation is confined to cancerous tissue, resulting in low toxicity and high specificity , thus addressing the dual challenges of "inadequate penetration" and "poor selectivity." In contrast, commonly used Cell Penetrating Peptides (CPPs) like TAT, although effective in transmembrane transport, lack tumor specificity and can lead to systemic toxicity .
Inspiration
Through our investigation, we identified the antimicrobial peptide BF2 and its engineered derivative, the cell-penetrating peptide BRⅡ. BRⅡ possesses three key characteristics :
No. 1
Delivery Capability:
BRⅡ is rich in arginine (R) and leucine (L), forming an optimal balance between moderate positive charge and hydrophobicity. This facilitates interaction with the anionic components of cancer cell membranes and promotes transmembrane transport. Literature supports its ability to deliver cargoes like EGFP into cells (A Cancer Specific Cell-Penetrating Peptide, BR2, for the Efficient Delivery of an scFv into Cancer Cells).
No. 2
Cancer Cell Specificity:
BRⅡ preferentially enters cancer cells via macropinocytosis, a targeting mechanism derived from the high expression of anionic gangliosides on the cancer cell surface. In contrast, commonly used cell-penetrating peptides (CPPs), while effective in transmembrane transport, lack tumor specificity and often lead to systemic toxicity .
No. 3
Reduced Toxicity:
Compared to the parent peptide BFⅡ, BRⅡ exhibits significantly reduced hemolytic toxicity at high concentrations, demonstrating improved biosafety .
Based on the unique properties of BRⅡ, we propose a technical strategy to engineer it into an Activatable Cell-Penetrating Peptide (ACPP). This designs a penetration delivery module for transporting therapeutic agents that otherwise cannot or have difficulty crossing membranes into intracellular targets to exert pharmacological effects. We connected BRⅡ to a negatively charged peptide segment via a matrix metalloproteinase MMP-9-sensitive linker, creating a "molecular switch" system. Concurrently, the anionic shielding significantly reduces non-specific uptake of BRⅡ in normal tissues. This design leverages the high expression of MMP-9 in the tumor microenvironment, ensuring that the ACPP is cleaved and activated specifically at the tumor site, releasing the membrane-penetrating and drug-carrying BRⅡ. This enables spatially specific drug delivery. This design further enhances specificity, successfully addressing the specificity deficiency caused by relying solely on electrostatic attraction between positive and negative charges—a deficiency that arises precisely from the charge interaction between the enriched positive charges of the peptide segment and the negatively charged gangliosides highly expressed on the surface of cancer cells. By overcoming the limitations of this single interaction mode, the biosafety of BRⅡ is effectively improved.
Furthermore, considering the long-term treatment needs for colorectal cancer, we designed a tumor-recognizing drug-releasing module for oral delivery via engineered bacteria. This aims to enable non-invasive, operator-independent administration with inherent gut targeting.
No. 1
Non-invasive and Operator-Independent:
Engineered bacteria are administered via enteric-coated capsules, requiring no skin penetration and featuring low operational complexity. This is particularly suitable for elderly patients or scenarios requiring long-term, regular dosing, enhancing patient adherence throughout extended treatment cycles.
No. 2
Inherent Gut Targeting:
Orally administered engineered bacteria reach the intestine and establish short-term colonization. The colonization ceases as the intestinal mucosa naturally renews itself. This approach is inherently suited for intestinal diseases and can effectively improve bioavailability compared to systemic circulation via methods like microneedle infusion.
Solution
Overview
Considering the actual physical conditions of colorectal cancer (CRC) patients, our solution — the "CRC-killer in and out" KIT (a delivery toolbox) — provides two administration methods: "In and ouT"
Highlights
Comprehensive Delivery Toolbox: "CRC-killer in and out" KIT integrates two administration methods: "In (biology - TRACER&Deliver)" and "ouT (hardware microneedle design)", providing a one - stop solution for patients with different needs.
Adaptable to Diverse Medical Scenarios: Capsules are convenient for self - medication at home, and intelligent microneedles are suitable for large - scale medical screening and treatment. The combination of the two can flexibly meet different scenarios such as home and public medical care, and relieve the pressure of medical resource allocation.
Patient - Centric Design: It offers diverse administration options, respects individual differences of patients, and improves treatment compliance. Meanwhile, the safe and efficient delivery methods reduce the dosage of drugs and the risk of potential side effects while ensuring efficacy.
In and Out: Biology
The TRACER protein comprises BRⅡ, a sensitive peptide, and a negative charged shielding peptide. In this study, interleukin-24 (IL-24) will be selected as the cargo of the activatable cell-penetrating peptide (ACPP). TRACER and IL-24 will be connected via an F Linker to construct the fusion protein TRACER-IL24, which will be used for subsequent functional verification.
- BRⅡ Peptide:
BRⅡ is a cancer-specific cell-penetrating peptide. Leveraging its positive charge, it preferentially engages in electrostatic interactions with gangliosides, which are highly expressed on cancer cell surfaces. This triggers lipid raft-mediated macropinocytosis, enabling efficient membrane penetration and the precise delivery of therapeutic molecules into the cytoplasm. Due to the lower density of anionic components on normal cell surfaces, the binding and internalization efficiency of BRⅡ in normal cells is significantly lower than in cancer cells. This characteristic allows BRⅡ to efficiently deliver therapeutic molecules specifically into cancer cells to exert their therapeutic effects.
- Anionic Shielding Peptide:
Connected to BRⅡ via the MMP-9-sensitive peptide, the anionic shielding peptide segment inhibits the penetrating activity of BRⅡ, preventing non-specific activation in non-tumor environments.
- Sensitive Peptide:
The PLGLAG sequence can be specifically cleaved by matrix metalloproteinase-9 (MMP-9). Based on this, through literature research combined with bioinformatics analysis, we found that the expression level of MMP-9 in colorectal cancer tissues is significantly higher than that in normal tissues, suggesting that MMP-9 has the potential to serve as a molecular marker for colorectal cancer.
- F Linker:
The F Linker is a flexible connecting sequence used to link two proteins, providing structural flexibility and functional cooperativity.
- IL-24 Protein:
IL-24 serves as the therapeutic drug molecule, possessing anti-cancer activity... Consequently, it achieves efficient, low-toxicity, multi-target, and broad-spectrum anti-cancer effects.
- It induces the expression of GADD family genes, phosphorylates eIF2α, activates caspase cascades, and induces apoptosis.
- It upregulates Bax/Bak, inhibits Bcl-2/Bcl-xL, promotes cytochrome c release, and induces mitochondria-dependent apoptosis.
- It blocks PI3K/AKT, STAT3, and Src signaling, inhibits VEGF/TGF-β, thereby suppressing tumor angiogenesis, cancer cell proliferation, and metastasis.
- It stimulates T/NK cells to secrete IFN-γ and TNF-α, enhancing immune recognition.
Consequently, it achieves efficient, low-toxicity, multi-target, and broad-spectrum anti-cancer effects against tumor cells.
BRⅡ possesses the ability to carry drugs into cells, relying on its positive charge to bind to cell membranes. The negatively charged shielding peptide inhibits the non-specific adsorption of BRⅡ in non-cancerous environments. The sensitive peptide is cleaved upon encountering MMP-9, which is highly expressed in the tumor microenvironment. IL-24 is connected to BRⅡ via a Flexible Linker. When the sensitive peptide is cleaved, the negatively charged peptide detaches, allowing BRⅡ to precisely deliver IL-24 into cancer cells, inducing their apoptosis.
In the intestinal tract of colorectal cancer patients, the level of oxidative damage is significantly elevated, primarily due to massive infiltration of inflammatory cells and mitochondrial dysfunction, leading to excessive production of reactive oxygen species (ROS) and reactive nitrogen species (RNS). These oxidants can directly attack DNA, inducing gene mutations. Based on this, we propose the:
This module utilizes the SOS promoter to sense DNA damage, activating the SRRz lysis protein to release the drug, while employing EGFP for visual monitoring of the process.
- Activation (Activator by DNA Damage):
In the high oxidative stress microenvironment of colorectal cancer, DNA damage products accumulate significantly. Therefore, we introduce the SOS response mechanism derived from E. coli: an inducible DNA repair system in E. coli that is activated when DNA is damaged. When bacterial DNA is damaged, the RecA protein is activated and mediates the auto-proteolysis of the LexA repressor, thereby derepressing the SOS promoter. The expression level from the SOS promoter increases, subsequently activating the downstream LuxI gene to express the acyl-homoserine lactone (AHL) synthase, completing the conversion from DNA damage to a biomolecular signal.
- Lysis:
When the intracellularly synthesized AHL accumulates to a certain threshold, it binds to the constitutively expressed LuxR protein, forming a LuxR-AHL complex. This complex specifically activates the Lux promoter, thereby driving the expression of the downstream lysis gene SRRz. SRRz encodes three lysis proteins: the holin encoded by the S gene forms pores in the cell membrane; the endolysin encoded by the R gene hydrolyzes the peptidoglycan layer of the cell wall; and the Rz gene product cleaves the linkages between the peptidoglycan and the outer membrane. These three proteins act synergistically to thoroughly disrupt the cell structure, ultimately leading to bacterial lysis.Once the to-be-lysed proteins are inactivated, the remaining bacterial population continues to grow and repeats the aforementioned process, forming an oscillatory drug delivery effect.
- TRACER-IL24::
An efficient fusion protein for anticancer therapy designed in this study comprises a TRACER domain and interleukin-24 (IL-24).
- Reporter:
The Lux promoter activates the expression of the EGFP reporter gene, used for characterizing the growth and lysis status of the engineered bacteria in vitro.
- Therapeutic:
Throughout this process, the strong constitutive J23100 promoterhttps://parts.igem.org/Part:BBa_J23100 continuously drives the expression of the therapeutic protein molecule IL-24. IL-24 is a protein molecule capable of inducing apoptosis in cancer cells. IL-24 is connected to TRACER via a Flexible Linker (Flinker), achieving efficient and precise anti-cancer functionality. This biological circuit design implements a comprehensive process of DNA damage detection and response, coupled with bacterial lysis and drug release.
Our TRACER and Deliver Module and Tumor-Sensing Drug Release Module work in concert to execute a coordinated "recognition-release-penetration-killing" process targeting cancer cells.
After the engineered bacteria reach the tumor site, the Tumor-Sensing Drug Release Module captures DNA damage signals caused by high oxidative stress via the SOS promoter. This activates the expression of the LuxI gene, leading to the synthesis of AHL. AHL then binds to the LuxR protein, forming a tetrameric complex that subsequently activates the Lux promoter. This activation drives the expression of the SRRz lysis proteins, resulting in bacterial lysis and the release of the IL-24 prodrug, which is constitutively expressed by the J23100 promoter.
Following drug release, the Activatable Cell-Penetrating Peptide (ACPP) delivery module takes over: BRⅡ, with its positive charge, specifically recognizes the relatively high abundance of gangliosides on the cancer cell surface. Concurrently, the anionic shielding peptide is linked to BRⅡ via an MMP-9-sensitive peptide linker. When matrix metalloproteinase-9 (MMP-9) activity is elevated in the microenvironment, the sensitive linker is cleaved, the shielding peptide dissociates, and BRⅡ is activated. The activated BRⅡ then carries IL-24 across the membrane into the cytoplasm. Inside the cell, IL-24 triggers multi-pathway synergistic apoptosis, achieving multi-target, efficient, and low-toxicity killing of the cancer cells.
- TRACER:
First, oral enteric-coated capsules with a calcium alginate shell are administered. These capsules gradually disintegrate in the intestine, releasing the engineered bacteria. These bacteria then colonize the intestinal mucosa and initiate the Tumor-Sensing and Drug Release Module. This module senses DNA damage within the high oxidative stress microenvironment of colorectal cancer via the SOS promoter, activating LuxI gene expression to produce AHL synthase. When AHL accumulates to a certain threshold, it binds to LuxR, triggering the Lux promoter to drive the expression of the SRRz lysis proteins, leading to bacterial lysis. Consequently, the IL-24 drug, constitutively expressed by the J23100 promoter, is released. Simultaneously, EGFP produces green fluorescence, allowing us to monitor the drug release status during in vitro experiments.
- Deliver:
Subsequently, the Penetration and Delivery Module comes into effect. The TRACER protein contains BRⅡ, the sensitive peptide, and the anionic shielding peptide. Within the tumor microenvironment, MMP-9 protease cleaves the sensitive peptide, causing the anionic shielding peptide to detach and thereby releasing active BRⅡ. Leveraging its positive charge, BRⅡ binds to the negatively charged gangliosides on the cancer cell surface, precisely delivering IL-24 into the cancer cell interior. Inside the cell, IL-24 triggers cancer cell apoptosis by modulating the synergistic effects of multiple signaling pathways, achieving highly efficient and low-toxicity anti-cancer activity.
- Lysis Cycle:
on our module, the engineered bacteria will exhibit a periodic oscillatory behavior of alternating growth and lysis in the intestinal tract. Within the patient's daily physiological rhythm, the interval between two meals serves as the core therapeutic window. During this period, the engineered bacteria undergo multiple rounds of lysis, releasing drugs continuously in a "pulsatile" manner to maintain therapeutic pressure on cancer cells.Meanwhile,the intestinal environment renewal (e.g., intestinal mucosa shedding and renewal) triggered by the patient's eating behavior will automatically terminate the current treatment cycle and clear the colonized engineered bacteria. This not only effectively prevents excessive proliferation of the bacterial population but also completes the "clearance" of colonization sites simultaneously, preparing for the implantation of the next batch of engineered bacteria and the initiation of a new round of treatment cycle, thereby ensuring the safety and sustainability of long-term treatment.
In and Out: Hardware
To address the challenges of patient acceptance and precise drug release, our hardware team developed a microneedle-based smart drug delivery system. Microneedle patches consist of tiny, dissolvable needles that painlessly penetrate the skin and release therapeutic proteins directly into the bloodstream, achieving higher delivery efficiency than traditional oral methods. To enable precise control over the release process, we integrated flexible electronics—including a heating film, PCB board, and smartphone-controlled circuits—into the patch. This design allows us to fine-tune the release rate and profile, supporting programmable modes such as rapid, delayed, or sustained delivery. The system can be used independently, offering a patient-friendly alternative to engineered bacteria, or in combination with them to provide adjustable therapeutic boosts when needed. By bridging synthetic biology with intelligent hardware, we created a dual therapeutic platform that is effective, controllable, and patient-centered.
References
- [1] Bray F, Laversanne M, Sung H, et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2024;74(3):229-263. doi: 10.3322/caac.21834.
- [2] Morgan E, Arnold M, Gini A, et al. Global burden of colorectal cancer in 2020 and 2040: incidence and mortality estimates from GLOBOCAN. Gut. 2023;72(2):338-344. doi: 10.1136/gutjnl-2022-327736.
- [3] Xia C, Dong X, Li H, et al. Cancer statistics in China and United States, 2022: profiles, trends, and determinants. Chin Med J (Engl). 2022;135(5):584-590. doi: 10.1097/CM9.0000000000002108.
- [4] Hernandez Dominguez O, Yilmaz S, Steele S R. Stage IV colorectal cancer management and treatment. J Clin Med. 2023;12(5):2072. doi: 10.3390/jcm12052072.
- [5] Dekker E, Tanis P J, Vleugels J L A, et al. Colorectal cancer. Lancet. 2019;394(10207):1467-1480. doi: 10.1016/S0140-6736(19)32319-0.
- [6] Alexander J L, Wilson I D, Teare J, et al. Gut microbiota modulation of chemotherapy efficacy and toxicity. Nat Rev Gastroenterol Hepatol. 2017;14(6):356-365. doi: 10.1038/nrgastro.2017.20.
- [7] Longley D B, Harkin D P, Johnston P G. 5-fluorouracil: mechanisms of action and clinical strategies. Nat Rev Cancer. 2003;3(5):330-338. doi: 10.1038/nrc1074.
- [8] Zhu Q, Chen Z, Paul P K, et al. Oral delivery of proteins and peptides: challenges, status quo and future perspectives. Acta Pharm Sin B. 2021;11(8):2416-2448. doi: 10.1016/j.apsb.2021.04.001.
- [9] Stintzing S, Modest D P, Rossius L, et al. FOLFIRI plus cetuximab versus FOLFIRI plus bevacizumab for metastatic colorectal cancer (FIRE-3): a post-hoc analysis of tumour dynamics in the final RAS wild-type subgroup of this randomised open-label phase 3 trial. Lancet Oncol. 2016;17(10):1426-1434. doi: 10.1016/S1470-2045(16)30269-8.
- [10] Sartore-Bianchi A, Trusolino L, Martino C, et al. Dual-targeted therapy with trastuzumab and lapatinib in treatment-refractory, KRAS codon 12/13 wild-type, HER2-positive metastatic colorectal cancer (HERACLES): a proof-of-concept, multicentre, open-label, phase 2 trial. Lancet Oncol. 2016;17(6):738-746. doi: 10.1016/S1470-2045(16)00150-9.
- [11] Misale S, Yaeger R, Hobor S, et al. Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature. 2012;486(7404):532-536. doi: 10.1038/nature11156.
- [12] Wilhelm S, Tavares A, Dai Q, et al. Analysis of nanoparticle delivery to tumours. Nat Rev Mater. 2016;1:16014. doi: 10.1038/natrevmats.2016.14.
- [13] Lizardo D Y, Kuang C, Hao S, et al. Immunotherapy efficacy on mismatch repair-deficient colorectal cancer: from bench to bedside. Biochim Biophys Acta Rev Cancer. 2020;1874(2):188447. doi: 10.1016/j.bbcan.2020.188447.
- [14] Ramos-Casals M, Brahmer J R, Callahan M K, et al. Immune-related adverse events of checkpoint inhibitors. Nat Rev Dis Primers. 2020;6:38. doi: 10.1038/s41572-020-0160-6.
- [15] Fujiwara Y, Kato S, Nesline M K, et al. Indoleamine 2,3-dioxygenase (IDO) inhibitors and cancer immunotherapy. Cancer Treat Rev. 2022;110:102461. doi: 10.1016/j.ctrv.2022.102461.
- [16] Tanaka H Y, Kano M R. Stromal barriers to nanomedicine penetration in the pancreatic tumor microenvironment. Cancer Sci. 2018;109(7):2085-2092. doi: 10.1111/cas.13630.
- [17] Petersson J, Schreiber O, Hansson G C, et al. Importance and regulation of the colonic mucus barrier in a mouse model of colitis. Am J Physiol Gastrointest Liver Physiol. 2011;300(2):G327-G333. doi: 10.1152/ajpgi.00422.2010.
- [18] Mortaja M, Cheng M M, Ali A, et al. Tumor-targeted cell-penetrating peptides reveal that monomethyl auristatin E temporally modulates the tumor immune microenvironment. Molecules. 2024;29(23):5618. doi: 10.3390/molecules29235618.
- [19] Lim K J, Sung B H, Shin J R, et al. A cancer specific cell-penetrating peptide, BR2, for the efficient delivery of an scFv into cancer cells. PLoS One. 2013;8(6):e66084. doi: 10.1371/journal.pone.0066084.
- [20] van Duijnhoven S M, Robillard M S, Nicolay K, Grüll H. Tumor targeting of MMP-2/9 activatable cell-penetrating imaging probes is caused by tumor-independent activation. J Nucl Med. 2011;52(2):279-286. doi: 10.2967/jnumed.110.082503.
- [21] Jiang T, Olson E S, Nguyen Q T, et al. Tumor imaging by means of proteolytic activation of cell-penetrating peptides. Proc Natl Acad Sci U S A. 2004;101(51):17867-17872. doi: 10.1073/pnas.0408191101.
- [22] Park C B, Kim H S, Kim S C. Mechanism of action of the antimicrobial peptide buforin II: buforin II kills microorganisms by penetrating the cell membrane and inhibiting cellular functions. Biochem Biophys Res Commun. 1998;244(1):253-257. doi: 10.1006/bbrc.1998.8159.
- [23] Din M O, Danino T, Prindle A, et al. Synchronized cycles of bacterial lysis for in vivo delivery. Nature. 2016;536(7614):81-85. doi: 10.1038/nature18930.
- [24] Leuzzi A, Grossi M, Di Martino M L, et al. Role of the SRRz/Rz1 lambdoid lysis cassette in the pathoadaptive evolution of Shigella. Int J Med Microbiol. 2017;307(4-5):268-275. doi: 10.1016/j.ijmm.2017.03.002.
- [25] Fisher P B. Is mda-7/IL-24 a “magic bullet” for cancer? Cancer Res. 2005;65(22):10128-10138. doi: 10.1158/0008-5472.CAN-05-3127.