How to start making phages for therapy

Issue 184 | July 15, 2022
23 min read
Capsid and Tail

This week Jessica Sacher takes us through her process for setting up a new phage-making system in the lab, including making sense of a dizzying array of options, managing overwhelm, and resisting ‘optimization creep’.

This week’s feature post is part of our new ‘Phage Australia Lab Series’, where we’re sharing what we’re doing at Phage Australia. Check out our last post, where Stephanie Lynch wrote about how we sent a phage around Australia and asked everyone to titre it!

Sponsor

Sponsor Capsid!

What’s New

News: LyseNTech, a Korean phage company led by Heejoon Myung, was named a leading research institute by the Korean Ministry of Trade, Industry and Energy. LyseNTech aims to develop ‘LNT103’, a phage-derived endolysin, as an inhaled treatment through a nebulizer.

Biotech news

Paper: Evading while invading: induction of CRISPR-Cas by mucin as a mean to escape phages adhered to mucus.

CRISPR-Cas SystemResearch paper

Paper: Bacteriostatic effects of phage F23s1 and its endolysin on Vibrio parahaemolyticus.

Antimicrobial developmentEndolysinsResearch paper

Pre-print: The defence island repertoire of the Escherichia coli pan-genome.

Genome annotationGenome sequencingPreprint

Pre-print: Temperate phage-antibiotic synergy across antibiotic classes reveals new mechanism for preventing lysogeny.

LysogenyPhage-antibiotic synergyPreprint

Latest Jobs

MicrobiomePhageomePost Doc
Post doc: The Pride Laboratory is seeking a Postdoctoral Fellow to investigate the role of bacteriophages as members of the human microbiome and to assist in the efforts to identify bacteriophages that kill human pathogens.
Marine phagesPhage TherapyPost DocViral ecology
Post doc positions: Multiple postdoctoral scientist positions (2+ years) are available in the School of Biological Sciences at the Georgia Institute of Technology in Atlanta to work on phage therapy & marine virus ecology.
Phage TherapyResearch AssociateSynthetic phages
Research associate: Armata is a clinical-stage biotechnology company focused on the development of precisely targeted phage therapeutics and is seeking a research associate to support the research and development effort by performing in vitro microbiological and associated microbiology/molecular biology testing/analysis.
Clinical researchPhage TherapyScientist
Infectious Diseases - Phage Scientist: The Division of Infectious Disease at the Mayo Clinic in Rochester, MN, is seeking scientist or physician-engaged in research with expertise or primary interest in phage therapy or phage basic science.
Infectious diseaseMicrobiologyResearch & DevelopmentScientific Director
Scientific Director: Janssen Biopharma Inc. is seeking an experienced and highly motivated Scientific Director to lead the discovery of novel antibacterial therapeutic agents, including phages.

Community Board

Anyone can post a message to the phage community — and it could be anything from collaboration requests, post-doc searches, sequencing help — just ask!

Drafting international guidelines for phage banks — initial brainstorming & organizational meeting

We would like to gather a working group that will draft suggested guidelines over the coming months. This brainstorming meeting is open to anyone in person or on Zoom (though please do not post the Zoom link on social media for security reasons), including those who already have experience managing phage banks and those who would like to initiate a phage bank.

When: Lunchtime (12:30 - 1:30) on Wednesday, July 20th (Portugal time) in Meeting Room #1 at Palacio Vila Flor (VoM2022 conference venue).

Any questions, contact Tobi Nagel: [email protected].

BiobankHybridMeeting

The Phage Bites symposium is back! With student/ECR talks and expert guest panelists Keith Shearwin, Stefanie Barbirz and Michael J. McDonald discussing #phage #adaptive #strategies. Free to attend! 2nd Aug 2022. Online. Register and submit an abstract.

Phage biologyVirtual Event

Jon Iredell (Director of Phage Australia) & Jessica Sacher (Phage Directory) will take part in an interactive ‘Meet the Researcher’ webinar hosted by the WIMR Foundation on phage therapy for AMR infections, and the work they’re doing at Phage Australia. This webinar is aimed at a public audience and all are welcome. Wed, 17 Aug 2022, 12.30pm–1.30pm, AEST.

Phage TherapyVirtual Event

Register to attend The Phage Canada Virtual Symposia 2022! This virtual event will be held August 17th - 19th and aims to promote the exchange of ideas within the phage field, provide a forum for students and postdocs to present their research and foster interactions between phage researchers. Deadline to register is Aug 13.

Virtual Event

The newly formed Belgian Society for Viruses of Microbes is excited to announce a 1-day inaugural symposium on Sept 23rd, 2022 in Leuven, Belgium!

In parallel, the BSVoM team is guest-editing a special issue in Viruses. They call on all Belgian scientists and their international collaborators to submit their related research in this upcoming issue.

New societySpecial IssueViruses of microbes

How to start making phages for therapy

Profile Image
Phage microbiologist and co-founder of Phage Directory
Co-founderPostdoctoral Researcher
Iredell Lab, Phage Directory, The Westmead Institute for Medical Research, Sydney, Australia, Phage Australia
Skills

Phage characterization, Phage-host interactions, Phage Therapy, Molecular Biology

I’m a co-founder of Phage Directory and have a Ph.D in Microbiology and Biotechnology from the University of Alberta (I studied Campylobacter phage biology). For Phage Directory, I oversee community building, phage sourcing, communications, science, and our awesome team of volunteers.

As of Feb 2022, I’ve recently joined Jon Iredell’s group in Sydney, Australia as a postdoctoral research scientist for the Phage Australia project. I’m diving back into the lab to help get Phage Australia’s country-wide phage therapy system up and running here, working to streamline workflows for phage sourcing, biobanking and collection of phage/bacteria/patient matching and monitoring data, and integrating it all with Phage Directory’s phage exchange, phage alerts and phage atlas systems. I’m also delving into phage manufacturing and quality control.

This article is part of the ‘Phage Australia Lab Series’, where we’ll be sharing the work we’re doing at Phage Australia. Check out our last post, where Stephanie Lynch wrote about how we sent a phage around Australia and asked everyone to titre it!

I’m writing this post partly for myself, to get my thoughts organized (I’m pretending you all are my virtual labmates!) but also, I think it could be helpful for others. I hope others see my process, send me helpful tricks / point out where I’m missing something, and hopefully get inspired to wrangle their own lab chaos and/or make phages for themselves!

Of note, Ali Khalid, PhD student in the Iredell Lab and Phage Australia team member, with the help of Helene Lebhar at UNSW’s Recombinant Products Facility, has done a huge amount of work getting our phage manufacturing off the ground. What follows is my perspective on getting myself organized so I can help extend Ali and Helene’s work to bridge some of our current gaps.

Patients are waiting on us

First, some context on why we need these phages, since of course, that will drive a lot of our decisions. At Phage Australia, we need phage. Not just any phage, but safe preps of phage we can use in patients. Here in Sydney, we’ve got three patients ready to be treated — they are just waiting for their phages.

The trouble is, we aren’t fully set up to make phages yet. We are still in the pilot phase of this work, and the funding for our own GMP phage-making setup that we can scale has yet to come in. Sure, we could get phages from other people, but the whole point of Phage Australia is to develop this capacity for ourselves.

While we wait for the grant (fingers crossed!), we need a system that works now so we can respond to cases that come in, and in the process, prepare to scale up and certify our process once we do get the funding.

Making phage is simple, right?

I came into this feeling pretty confident I could make this work. There are tons of ways you can make phages. Tons of papers written about it. Lots of people make phages. You just need a flask, right? So I set out to find a phagemaking protocol. My strategy was to pick a few groups I know are making phages regularly, to roughly the standards we need (US FDA, emergency/compassionate use), check out their most recent papers, and do what they do.

Quickly I realized everyone does things slightly differently. Ok, no problem, I thought, I’ll test a few things and whatever works best, and is easiest, that’s what I’ll do. But what does ‘best’ mean? What does ‘easiest’ mean? Which steps can I skip? Which pitfalls do I want to avoid? I descended down a rabbit hole of things that could matter. It seemed like each paper highlighted why their method was better than the others. And I didn’t have the materials to try everything. Or the time. Suddenly, this did not feel like a simple task. I felt like I had a thousand decisions to make. For instance, how much phage to cell ratio do you use? When do you add phage? How long do you let it incubate? Will more aeration be good for the phage or bad? Should I spin down after 5 hours, or wait until the next day?

And that’s just making the phage. Then there’s purifying it. Some use cesium chloride, while others say that’s an old, crude method that brings in dangerous chemicals. And it’s a pain to set up — careful not to disturb the gradient! Others use organic solvents, or ultrafiltration… Cytiva, Sartorius, Akta Flux, ion exchange, polishing, buffer exchange. So many new words to learn, each with specifications and pore sizes and capacities.

Case in point, I quickly went from ‘this is straightforward’ to option paralysis. What I thought was an opportunity (I get to design this from the ground up! No biases! First principles!) started becoming a dizzying array of options, with no place to start.

Whiteboard
A snapshot of what my brain was trying to keep track of as I thought about how to get a minimally functional phage-making system up and running.

Start with the end in mind (figure out what we’re optimizing for)

To give myself somewhere to start, I took a breath and thought, ok, what are the actual things we need? What are we optimizing for? What are our constraints?

To address our imminent patient needs, our needs are actually simple. We need enough phages, and we need them to have low endotoxin to be safe. And right now, we have limited time and materials.

Our needs

  • Enough phages
  • Safe phages

Our constraints

  • Limited labour time
  • Limited ability to order new equipment

What is enough?

Phage Australia has a protocol for phage therapy called STAMP (recently approved and started recruiting/treating patients earlier this year). In it, dosing is standardized: 10^9 PFU per dose. Minimum 28 courses of that. So we need minimum 2.8 x 10^10 phages for each patient. Let’s aim for some wiggle room, and try to get 10^12 phages per patient.

What is safe?

We know how low the endotoxin content needs to be (FDA has guidelines, and Australia will likely follow this: you can only put a max of 5 endotoxin units per kg/h into a patient’s vein). So that means we’re successful if we can get below maximum endotoxin concentration (5 EU/kg/h, meaning for a 70-kg patient, we should aim for below 350 EU per phage dose).

Limited personnel and equipment

We only have two postdocs devoting part of their time to this, so phage-making should be doable in about 20 person-hours or less per week. And since we’re in a relatively isolated country in the middle of a global supply chain crisis, trying to stretch the grant dollars we have until the end of the year, let’s try to limit ourselves to things we have in the lab, and avoid ordering anything new.

Decision: let’s optimize for making enough safe phages easily, using what we already have.

Identify the core components of the work

At its core, we need to make a bunch of phages, and remove the endotoxin. Next let’s break it down further. What does each step entail? How do we know if it’s successful?

1. Phage production

  • Grow phage and cells together
  • Separate out the phage

Successful if: we achieve our minimum per-batch phage amount (10^12 PFU)

2. Endotoxin removal

  • Concentrate the sample
  • Remove the endotoxin
  • Get rid of anything dangerous you added to get rid of the endotoxin

Successful if: we get below maximum endotoxin concentration (5 EU/kg/h, ie. 350 EU per 10^9 PFU)

Note: endotoxin removal is more than just endotoxin removal

I have identified at least two shadow steps that come before and after the actual removal of endotoxin: concentrating your sample beforehand, and cleaning it after. Most endotoxin removal steps I’ve looked at require a concentrated phage solution before moving to endotoxin removal. For example, CsCl gradients call for adding ~3 mL phages, meaning no matter how much phage you make, you need it to be concentrated into 3 mL if you want to use CsCl. Another option is octanol — for this step, you’re adding octanol approximately 1:1 to your phage. Unless you want to go through litres of octanol, you’ll want to concentrate your phage ahead of time. Then, after endotoxin removal, you need to remove whatever you added to remove the endotoxin that might be toxic (e.g. cesium, octanol).

Look at our options for each step

For each step, there are many ways you can do it. I found it helpful to list the options/considerations for each. I found ideas by reading a few papers from friends in the phage field I know make phages for compassionate use cases, like TAILOR, IPATH, and the Phage on Tap method, developed by one of our own Phage Australia colleagues.

Decide what to do at each step, and document why

Next: deciding what to do for each step. To illustrate my thought process and how/why I’m making these decisions, I started laying this out in a table, along with what I’ve tried to date. For each step, I picked one approach (the easiest option available).

Table of Options & Processes

Minimize optimization creep!

Even though I thought I knew what I was optimizing for, I found that my instinct was to get into the lab and start optimizing everything, setting up little parallel experiments here and there ‘just to see’. I found that I was constantly trying to optimize for the highest titre, even though that is NOT what I decided to optimize for. It took me a while to realize this was happening, and I still have to catch myself constantly and resist that urge.

I came to realize that while there’s a lot we COULD optimize, we shouldn’t try to do this. It would make the task at hand take forever, most likely overwhelm us, and ultimately not help us achieve our goals (patients getting enough safe phages fast!). Hence why I found it helpful to get very clear on what I was optimizing for, then look at all my options through that lens. Given I decided we’re going for ‘good enough’, (ie. enough phages, safe enough), that means if a step meets that minimum (ie. we look like we’re on track to hit at least 10^12 phages at the end), we move on. Of course, if in the future we use a phage that doesn’t propagate well, or purify well, then that would be grounds to start tweaking the steps prior to that point. Otherwise, we’ll move along.

Of note, most of my work so far has been with two phages, Pae7, a Pseudomonas phage, and Ec134B, an E. coli phage. I fully anticipate things to change with different phages. I’m hoping we can at least find a baseline that works pretty well for lots of phages, then tweak from there when we need to (and only then).

A few more notes, learnings, questions

Getting high enough titres

For a while I wanted to achieve higher titres so I could skip or reduce the need for a concentration step prior to endotoxin removal (I’d heard it could be an annoying bottleneck). I played with a bunch of temperature, flask, time and media parameters, which didn’t affect production by much (max about half a log). For now I’ve deemed most these kinds of tweaks to the growth step not worth doing.

For example, leaving cells+phage together for the weekend at 4C before filtering out the cell debris led to 2-4x more phage, which I felt wasn’t enough of a jump to justify the extra time and unknown effects on the phage that that extra step would bring. Going forward I think we should just focus on choosing a good host strain, letting the phages grow to the titres they get to using standard conditions for that strain, and being ok with having a concentration step.

If phages are finicky, switching to solid media is something I think will help a lot to increase titres (we haven’t gone there so far, since others in our group have seen agar cause problems downstream, but I still think this could be a reasonable tradeoff if the phages won’t propagate well in liquid).

The one thing that worked the best to improve titres was changing out the propagation strain (led to about a log increase). Based on this, I think doing a host range panel to make sure we’re using the best strain we have for that phage would be worth the effort.

Getting rid of endotoxin

So far I’m happy with the amount of effort that octanol required (other than the dialysis step which I’d like to replace, since it was fairly laborious and used litres of ethanol). It will be interesting to see if it’s sufficient to remove endotoxin without removing too much phage. Jonas Van Belleghem and colleagues have done a really nice study contrasting different endotoxin removal methods, and octanol seems to be a reasonable choice (though it of course depends on the phage). (We have a large amount in the lab right now, and it only took a few mLs to do an extraction, though I’ll need to check how much it is to replace).

Beyond that, I want to try CsCl; first, gradientless (I only recently found out you don’t always have to build a gradient, thanks to Sabrina Green at TAILOR!) (her Youtube video and protocol here). The trouble is this method calls for a refractometer to measure density, but we don’t have one. I may try without (I’ve also found this protocol by the Center for Phage Technology which seems to do this). If that doesn’t work, I’ll do the gradient. Who knows though, we may get our Akta Flux (along with some chromatography columns), before I have the chance or the need to try CsCl.

Concluding thoughts

Thanks for reading! I hope you can take away from this that a complex array of options in the lab can be simplified into a path forward. The way I’ve found is helpful is:

  1. Decide what you’re optimizing for (what would success look like?)
  2. Break the project into its core components
  3. For each core component, make a list of what your options are
  4. Filter for materials you have access to
  5. Make a choice of one thing to try first for each step (and document why!)

Ultimately you should have one thing to try for each core component of your process. I decided not to order anything new, then picked what seemed like the easiest thing of the options I had available. I resisted the urge to do everything in parallel and compare based on which gave the highest titre (I was only able to resist this urge once I got clear on what I was optimizing for: hitting a minimum phage titre, the easiest way possible; NOT maximizing phage titre). I found it helped to make a table, then to force myself to list the core function of the step, the options I had available, the decision of what to try first for that step, the outcome, and the next step.

What felt before like a dizzying mess now feels like a framework I can build on. As I come across new options, I can look at my table and ask myself, is this new option going to be less laborious or ‘better’ than what I’m already doing? What evidence do I have for that? Importantly, is it ‘better’ according to my predefined success metric, or am I starting to optimize additional things I don’t need to optimize? (Beware optimization creep!)

Anyway, I’m happy with this framework for now — I’ll see how it serves me! Would love to hear others’ thoughts, helpful methods you’ve used to manage complexity in the lab and avoid option paralysis, as well as any specifics or magical phage-making tricks I’ve missed here!

Resources that helped me

Acknowledgments

  • Stephanie Lynch, my co-pilot here in the Iredell lab — we’re both closely working on all of this, trying to navigate doing our work as a team rather than as individuals; I could not imagine a better lab co-pilot!
  • Ali Khalid, who’s been working on purification of phage with Phage Australia for the last several months, and has helped me get up to speed on what it can entail. I’ve learned a ton from what he’s done, and he’s helped me get this all straight in my head.

Thanks to you readers for being my virtual labmates!

My dream would be that scientists everywhere would blog about the work they’re doing, the decisions they’re making and why, so we can all learn from each other’s thinking and experience. In my mind there’s no good reason not to. It’s not like one’s blog musings about why they ultracentrifuged at one speed vs. another are going to prevent journals from accepting our papers.

If we’re worried about looking stupid, well, I guess that’s the only risk left. I definitely was when we started Capsid & Tail 3.5 years ago. But as I’ve found from putting out the newsletter/blog every week since, and getting told exactly zero times that I’m stupid, I’ve realized it’s probably not that big a risk to say things online, without the shield of peer review and co-authors. You should try it! (Conveniently, we have a guest writer program for C&T!)

For more on why you should share what you’re working on as you go, check out this great article on how publishing your thoughts increases your luck. It inspired me to write this this week!

Capsid & Tail

Follow Capsid & Tail, the periodical that reports the latest news from the phage therapy and research community.

We send Phage Alerts to the community when doctors require phages to treat their patient’s infections. If you need phages, please email us.

Sign up for Phage Alerts

In collaboration with

Mary Ann Liebert PHAGE

Supported by

Leona M. and Harry B. Helmsley Charitable Trust

Crossref Member Badge